You are on page 1of 40

EVMS COVID-19 MANAGEMENT PROTOCOL

An overview of the MATH+ and I-MASK+ Protocols


Developed and Updated by Paul Marik, MD, FCP (SA), FRCP (C), FCCP, FCCM.
Professor of Medicine, Chief of Pulmonary and Critical Care Medicine
Eastern Virginia Medical School
December 17th, 2020

This is our recommended approach to COVID-19 based on the best (and most recent) literature. This is a
highly dynamic topic; therefore, we will be updating the guideline as new information emerges.
EVMS COVID website: https://www.evms.edu/covid-19/medical_information_resources/
Short url: evms.edu/covidcare

Disclaimer: The information in this document is provided as guidance to physicians World-Wide


on the prevention and treatment of COVID-19. Our guidance should only be used by medical
professionals in formulating their approach to COVID-19. Patients should always consult with
their physician before starting any medical treatment.

Page 1 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 1. The course of COVID-19 and General Approach to treatment

THIS IS A STEROID RESPONSIVE DISEASE:


HOWEVER, TIMING IS CRITICAL

Page 2 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Table 1. Pharmacological therapy for COVID by stage of illness: What has worked and what
has failed*

*based on randomized controlled trials (see supporting information below)

Page 3 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 2. Timing of the initiation of anti-inflammatory therapy

Page 4 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 3. Time course of laboratory tests for COVID-19

Figure 4. SARS-Co-V-2 RNA genome

Page 5 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


It should be noted that there is no cure or “Magic-bullet” for the prevention or treatment of COVID-19.
However, recently, a number of therapeutic agents have shown promise for both the prevention and
treatment of COVID-19 including ivermectin, Vitamin D, quercetin, melatonin and corticosteroids.
Furthermore, it is likely that no single drug will be effective in treating this complex disease and that
multiple drugs with different mechanisms of action and used in specific phases of the disease will be
required.

Prophylaxis
While there is no “Level 1 evidence” that this “cocktail” will prevent/mitigate against COVID-19 we
believe there is significant evidence supporting the efficacy of the individual agents included in the
prophylactic protocol. This protocol MUST be part of an overall strategy which includes common sense
public health measures, i.e. masks, social distancing, and avoidance of large groups of people.
Furthermore, it should be noted that there is emerging evidence suggesting that IVERMECTIN may be
highly effective in the prevention and treatment of COVID-19. It is important to emphasize that ALL of
the medications included in our prophylactic regimen are inexpensive, safe, and widely available.
• Vitamin D3 1000–3000 IU/day. Note RDA (Recommended Daily Allowance) is 800–1000 IU/day.
The safe upper-dose daily limit is likely < 4000 IU/day. [1-22] Vitamin D insufficiency has been
associated with an increased risk of acquiring COVID-19 and from dying from the disease.
Vitamin D supplementation may therefore prove to be an effective and cheap intervention to
lessen the impact of this disease, particularly in vulnerable populations, i.e. the elderly, those of
color, obese and those living > 45o latitude. [7-22]
• Vitamin C 500 mg BID (twice daily) and Quercetin 250 mg daily. [23-34] It is likely that vitamin C
and quercetin have synergistic prophylactic benefit. [35] It should be noted that in vitro studies
have demonstrated that quercetin and other flavonoids interfere with thyroid hormone
synthesis at multiple steps in the synthetic pathway. [36-39] The use of quercetin has rarely
been associated with hypothyroidism. The clinical impact of this association may be limited to
those individuals with pre-existent thyroid disease or those with sub-clinical thyroidism.[40] In
women high consumption of soya was associated with elevated TSH concentrations.[41] The
effect on thyroid function may be dose dependent, hence for chronic prophylactic use we
suggest that the lowest dose be taken. Quercetin should be used with caution in patients with
hypothyroidism and TSH levels should be monitored. It should also be noted quercetin may have
important drug-drug interactions; the most important drug-drug interaction is with cyclosporin
and tacrolimus. [42] In patients taking these drugs it is best to avoid quercetin; if quercetin is
taken cyclosporin and tacrolimus levels must be closely monitored.
• Melatonin (slow release): Begin with 0.3 mg and increase as tolerated to 2 mg at night. [43-50]
• Zinc 30–50 mg/day (elemental zinc). [23,30,32,33,51-55]
• B complex vitamins [56-60]
• Ivermectin for postexposure prophylaxis (see ClinTrials.gov NCT04422561). 0.2 mg/kg (12 mg)
immediately then repeat day 3.
• Ivermectin for pre-exposure prophylaxis (in HCW) and for prophylaxis in high-risk individuals
(> 60 years with co-morbidities, morbid obesity, long term care facilities, etc). 0.15–0.2 mg/kg
Day 1, Day 3 and then weekly for 10 weeks, followed by biweekly dosing. [5,61-64] (also see
ClinTrials.gov NCT04425850). See dosing Table below. NB. Ivermectin has a number of
potentially serious drug-drug interactions. Please check for potential drug interaction at
Ivermectin Drug Interactions - Drugs.com. The most important drug interactions occur with
cyclosporin, tacrolimus, anti-retroviral drugs, and certain anti-fungal drugs.

Page 6 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


• Optional: Famotidine 20–40 mg/day [55–61]. Low level evidence suggests that
famotidine may reduce disease severity and mortality. However, the findings of some
studies are contradictory. While it was postulated that famotidine inhibits the SARS-
CoV-2 papain-like protease (PLpro) as well as the main protease (3CLpro) this
mechanism has been disputed.[58] Furthermore, a single study suggested that users of
PPI’s had a significantly increased odds for reporting a positive COVID-19 test when
compared with those not taking PPIs, while individuals taking histamine-2 receptor
antagonists were not at elevated risk.[62] This data suggest that famotidine may be the
drug of choice when acid suppressive therapy is required.
• Optional/Experimental: Interferon-α nasal spray for health care workers [54]

Ivermectin dosing: 150-200 ug/kg or fixed dose of 12 mg (≤ 80kg) or 18 mg (≥ 80kg).[65] Depending on


the manufacturer ivermectin is supplied as 3mg, 6 mg or 12 mg tablets.
50-64.9 kg - 12mg
65-79.9 kg - 15mg
80-94.9 kg - 18mg
95-109.9 kg - 21mg
≥ 110 kg - 24mg

Symptomatic patients at home (for the duration of acute symptoms)


• Vitamin C 500 mg BID and Quercetin 250–500 mg BID
• Zinc 75–100 mg/day (elemental zinc)
• Melatonin 10 mg at night (the optimal dose is unknown) [50]
• Vitamin D3 2000–4000 IU/day. Calcifediol 0.2 mg is an alternative. [66]
• Highly recommended: Ivermectin 0.15–0.2 mg/kg orally (repeat on day 3). [1-5,63,67-78] See
Table 1, Figure 5 and ClinTrials.gov NCT04523831. See drug-drug interactions above.
• ASA 81–325 mg/day (unless contraindicated). ASA has antiinflammatory, antithrombotic, and
antiviral effects.[79,80] Platelet activation may play a major role in propagating the
prothrombotic state associated with COVID-19. [81]
• B complex vitamins
• Optional: Famotidine 40 mg BID (reduce dose in patients with renal dysfunction) [82-88].
• Optional: Vascepa (Ethyl eicosapentaenoic acid) 4g daily or Lovaza (EPA/DHA) 4g daily;
alternative DHA/EPA 4g daily. Vascepa and Lovaza tablets must be swallowed and cannot be
crushed, dissolved or chewed. Omega-3 fatty acids have anti-inflammatory properties and play
an important role in the resolution of inflammation. In addition, omega-3 fatty acids may have
antiviral properties. [32,89-92]
• Optional: Interferon-α/β s/c, nasal spray or inhalation. [93-96] It should be noted that Zinc
potentiates the effects of interferon.[97,98]
• In symptomatic patients, monitoring with home pulse oximetry is recommended (due to
asymptomatic hypoxia). The limitations of home pulse oximeters should be recognized, and
validated devices are preferred.[99] Multiple readings should be taken over the course of the
day, and a downward trend should be regarded as ominous.[99] Baseline or ambulatory
desaturation < 94% should prompt hospital admission. [100] The following guidance is
suggested: [99]

Page 7 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


o Use the index or middle finger; avoid the toes or ear lobe
o Only accept values associated with a strong pulse signal
o Observe readings for 30–60 seconds to identify the most common value
o Remove nail polish from the finger on which measurements are made
o Warm cold extremities prior to measurement
• Not recommended: Hydroxychloroquine (HCQ). The use of HCQ is extremely controversial.[101]
The best scientific evidence to date suggests that HCQ has no proven benefit for post exposure
prophylaxis, for the early symptomatic phase and in hospitalized patients. [102-120] Considering
the unique pharmacokinetics of HCQ, it is unlikely that HCQ would be of benefit in patients with
COVID-19 infection (it takes 5–10 days to achieve adequate plasma and lung
concentrations).[112,121-123] Finally, it should be recognized that those studies which are
widely promoted to support the use of HCQ are severely methodologically flawed.[124-127]
• Not recommended: Systemic or inhaled corticosteroids (budesonide). In the early symptomatic
(viral replicative phase), corticosteroids may increase viral replication and disease severity.[128]
An OpenSAFELY analysis in patients with COVID-19 demonstrated a higher risk of death in COPD
and asthmatic patients using high dose ICS. [129] The role of ICS in the pulmonary phase is
unclear as patients require systemic corticosteroids to dampen the cytokine storm, with ICS
having little systemic effects.

Page 8 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Mildly Symptomatic patients (on floor/ward in hospital):

• Vitamin C 500–1000 mg q 6 hourly and Quercetin 250–500 mg BID (if available)


• Zinc 75–100 mg/day
• Melatonin 10 mg at night (the optimal dose is unknown) [50]
• Vitamin D3 20,000–60,000 IU single oral dose. Calcifediol 0.2–0.5 mg is an alternative. [66] This
should be followed by 20,000 IU D3 (or 0.2 mg calcifediol) weekly until discharged from hospital.
Calcifediol is more efficiently absorbed, achieves 25-OH vitamin D levels quicker and is three
times more potent than vitamin D3. [130,131] However, it is important to note that the optimal
dose of vitamin D in the acute setting is unknown.[132,133] Very high doses may paradoxically
block the vitamin D receptor.
• Highly recommended: Ivermectin 0.15–0.2 mg/kg orally (12 mg) and repeat on day 3 [1-5,63,67-
77]. It should be noted that ivermectin has potent anti-inflammatory properties apart from its
antiviral properties.[134-136] See Table 1 and Figure 5. See drug-drug interaction above.
• Enoxaparin 60 mg/day [75,137-150] Consider increasing the dose to 1mg/kg q 12 hourly in those
with a high D-Dimer or an increasing D-Dimer (see Xa monitoring below).
• ASA 325 mg (if not contraindicated). Moderate-severe COVID infection results in profound
platelet activation contributing to the pro-thrombotic state and increasing the inflammatory
response.[151-153]
• Methylprednisolone 40 mg q 12 hourly; increase to 80 mg and then 125 mg q 12 hourly in
patients with progressive symptoms and increasing CRP. There is now overwhelming and
irrefutable evidence that corticosteroids reduce the risk of death in patients with the pulmonary
phase of COVID-19 i.e. those requiring supplemental oxygen or higher levels of support. [154-
166] The role of inhaled corticosteroids (budesonide) is unclear and appears to be rather
limited.
• B complex vitamins
• Optional: Famotidine 40 mg BID (20–40 mg/day in renal impairment). [82-88]
• Optional: Vascepa (Ethyl eicosapentaenoic acid) 4g daily or Lovaza (EPA/DHA) 4g daily;
alternative DHA/EPA 4g daily.
• Optional (not recommended): Remdesivir 200 mg IV loading dose D1, followed by 100mg day IV
for 9 days. [167,168] This agent has been reported to reduce time to recovery (based on an
ordinal scale) in patients requiring low levels of supplemental oxygen. [168,169] The recently
published SOLIDARITY trial demonstrated no mortality benefit of this agent in the entire
treatment cohort or any subgroup.[170] Considering the high cost of this agent and the lack of
benefit on patient centered outcomes the role of this drug seems very limited.
• N/C 2L/min if required (max 4 L/min; consider early t/f to ICU for escalation of care).
• Avoid Nebulization and Respiratory treatments. Use “Spinhaler” or MDI and spacer if required.
• T/f EARLY to the ICU for increasing respiratory signs/symptoms, increasing oxygen requirements
and arterial desaturation.

Page 9 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Progressive Respiratory symptoms (hypoxia- requiring N/C ≥ 4 L min: admit to ICU):
Essential Treatment (dampening the STORM); MATH + [171]
1. Methylprednisolone 80 mg loading dose then 40 mg q 12 hourly for at least 7 days and until
transferred out of ICU. In patients with an increasing CRP or worsening clinical status increase the
dose to 80 mg q 12 hourly (then 125mg q 12 hourly), then titrate down as appropriate. [154-166]
Pulse methylprednisolone 250–500 mg mg/day may be required.[164] As depicted in Table 1,
methylprednisolone is the corticosteroid of choice.
2. Ascorbic acid (Vitamin C) 3g IV q 6 hourly for at least 7 days and/or until transferred out of
ICU.[27,172-183]. Note caution with POC glucose testing (see below). Oral absorption is limited by
saturable transport and it is difficult to achieve adequate levels with PO administration. Should IV
Vitamin C is not available, attempts should be made to administer PO vitamin C at a dose of 1g
every 4–6 hours. Mega-dose vitamin C should be considered in severely ill patients and as salvage
therapy: 25g vitamin C in 200-500 cc saline over 6 hours daily for 4 days, then 3g IV q 6 hourly for
total of 7-10 days of treatment. [184] (also see https://www.youtube.com/watch?v=Au-
mp6RZjCQ )
3. Full anticoagulation: Unless contraindicated we suggest FULL anticoagulation (on admission to
the ICU) with enoxaparin, i.e., 1 mg kg s/c q 12 hourly (dose adjust with Cr Cl < 30mls/min). There
is now good evidence that high intensity anticoagulation reduces mortality of hospitalized
patients with COVID-19. [137,139,140,142-150,185] Heparin is suggested with CrCl < 15 ml/min.
Due to augmented renal clearance patients may have reduced anti-Xa activity despite standard
dosages of LMWH.[186] We therefore recommend monitoring anti-Xa activity in underweight and
obese patients, those with chronic renal failure and in those patients with an increasing D-dimer,
aiming for an anti-Xa activity of 0.6–1.1 IU.ml.

Note: A falling SaO2 and the requirement for supplemental oxygen should be a trigger to start anti-
inflammatory treatment (see Figure 2).

Note: Early termination of ascorbic acid and corticosteroids will likely result in a rebound effect with
clinical deterioration (see Figure 6).

Additional Treatment Components (the Full Monty)

4. Highly recommended: Ivermectin 0.15–0.2 mg/kg orally and repeat on day 2 (see doing above).
[1-3,67,70-78,134-136,187-193] Note that ivermectin has potent antiviral and ant-inflammatory
effects. See Table 1 and Figure 5.
5. Melatonin 10 mg at night (the optimal dose is unknown).
6. Calcifediol 0.2–0.5 mg. [66] This should be followed by 0.2 mg calcifediol weekly until discharged
from hospital. Vitamin D3 takes many days to be converted to 25OH vitamin D; [194] this may
explain the lack of benefit of D3 in patients hospitalized with severe COVID-19. [195]
7. Thiamine 200 mg IV q 12 hourly [196-201] Thiamine may play a role in dampening the cytokine
storm. [197]
8. ASA 325 mg. COVID infection results in profound platelet activation contributing to the severe
pro-thrombotic state and increasing the inflammatory response.[151-153] As the risk of
significant bleeding is increased in patients receiving both ASA and heparin, ASA should therefore
not be used in patients at high risk of bleeding. In addition (as noted below) patients should
receive famotidine concurrently.
9. B complex vitamins
10. Magnesium: 2 g stat IV. Keep Mg between 2.0 and 2.4 mmol/l. [59] Prevent hypomagnesemia
(which increases the cytokine storm and prolongs Qtc). [202-204]

Page 10 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


11. Optional: Doxycycline 100mg daily for 5 days. Doxycycline is a broad-spectrum antibiotic which
has synergistic anti-viral and anti-inflammatory effects when combined with Ivermectin.
[6,68,73,205]
12. Optional (Consider in severe cases). Anti-serotonin agents. Platelet activation results in the release
of serotonin, which may contribute to the “cytokine storm”. [206]Therefore, the serotonin
receptor blocker cyproheptadine 4–8 mg PO q 6 hours should be considered.
13. Optional. Atorvastatin 80 mg/day. Statins have pleotropic anti-inflammatory, immunomodulatory,
antibacterial, and antiviral effects. In addition, statins decrease expression of PAI-1. Simvastatin has
been demonstrated to reduce mortality in the hyper-inflammatory ARDS phenotype. [207]
Preliminary data suggests atorvastatin may improve outcome in patients with COVID-19.[208-212]
Due to numerous drug-drug interactions simvastatin should be avoided.
14. Optional: Famotidine 40 mg BID (20–40 mg/day in renal impairment). [82-88].
15. Optional: Vascepa, Lovaza or DHA/EPA 4g day (see above).
16. Not recommended: The role of azithromycin in the treatment of COVID-19 is controversial. The best
information to date suggests that azithromycin is of little benefit.[213,214]
17. Not recommended: Remdesivir. Has not benefit at this stage of the disease.
18. Not recommended. Convalescent serum [215,216] or monoclonal antibodies. (Eli Lilly recently
announced that they are suspending their ACTIV-33 clinical trial as their monoclonal antibody failed
to demonstrate a clinical benefit in hospitalized patients).
19. Not recommended. Tocilizumab. Four RCTS have now failed to demonstrate a clinical benefit from
tocilizumab. [217-220]
20. Broad-spectrum antibiotics if superadded bacterial pneumonia is suspected based on procalcitonin
levels and resp. culture (no bronchoscopy). Due to the paradox of hyper-inflammation and immune
suppression (a major decrease of HLA-DR on CD14 monocytes and T cell dysfunction) secondary
bacterial and fungal infection is not uncommon. [221]
21. Maintain EUVOLEMIA (this is not non-cardiogenic pulmonary edema). Due to the prolonged
“symptomatic phase” with flu-like symptoms (6–8 days) patients may be volume depleted. Cautious
rehydration with 500 ml boluses of Lactate Ringers may be warranted, ideally guided by non-
invasive hemodynamic monitoring. Diuretics should be avoided unless the patient has obvious
intravascular volume overload. Avoid hypovolemia.
22. Early norepinephrine for hypotension. It should however be appreciated that despite the cytokine
storm, vasodilatory shock is distinctly uncommon in uncomplicated COVID-19 (when not
complicated by bacterial sepsis). This appears to be due to the fact that TNF-α which is “necessary”
for vasodilatory shock is only minimally elevated.
23. Escalation of respiratory support (steps); Try to avoid intubation if at all possible, (see Figure7)
• Accept “permissive hypoxemia” (keep O2 Saturation > 84%); follow venous lactate and
Central Venous O2 saturations (ScvO2) in patents with low arterial O2 saturations
• N/C 1–6 L/min
• High Flow Nasal canula (HFNC) up to 60–80 L/min
• Trial of inhaled Flolan (epoprostenol)
• Attempt proning (cooperative repositioning-proning) [222,223]
• Intubation … by Expert intubator; Rapid sequence. No Bagging; Full PPE. Crash/emergency
intubations should be avoided.
• Volume protective ventilation; Lowest driving pressure and lowest PEEP as possible. Keep
driving pressures < 15 cm H2O.
• Moderate sedation to prevent self-extubation
• Trial of inhaled Flolan (epoprostenol)
• Prone positioning.

Page 11 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


There is widespread concern that using HFNC could increase the risk of viral transmission. There is
however, no evidence to support this fear. HFNC is a better option for the patient and the health
care system than intubation and mechanical ventilation. CPAP/BiPAP may be used in select
patients, notably those with COPD exacerbation or heart failure.

A sub-group of patients with COVID-19 deteriorates very rapidly. Intubation and mechanical
ventilation may be required in these patients.

Table 2: Comparison of Methylprednisolone, Dexamethasone and Hydrocortisone- Number


Need to Treat (NNT)

Page 12 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 5. Metaanalysis of Ivermectin clinical studies (in hospital mortality)

24. Salvage Treatments


• High dose bolus corticosteroids; 250–500 mg/day methylprednisolone [162,164] Plasma
exchange [224-230]. Should be considered in patients with progressive oxygenation failure
despite corticosteroid therapy as well as in patients with severe MAS. Patients may require
up to 5 exchanges. FFP is required for the exchange; giving back “good humors” appears to
be more important than taking out “bad humors”.
• Mega-dose vitamin C should be considered in severely ill patients and as salvage therapy: 25g
vitamin C in 200-500 cc saline over 6 hours daily for 4 days, then 3g IV q 6 hourly for total of
7-10 days of treatment. [181] (also see https://www.youtube.com/watch?v=Au-mp6RZjCQ)
• In patients with a large dead-space ventilation i.e. high PaCO2 despite adequate minute
ventilation consider “Half-dose rTPA” to improve pulmonary microvascular blood flow; 25mg
of tPA over 2 hours followed by a 25mg tPA infusion administered over the subsequent 22
hours, with a dose not to exceed 0.9 mg/kg followed by full anticoagulation.[231,232]
• ECMO [233,234]. Unlike “typical ARDS” COVID-19 patients do not progress into a resolution
phase. Rather, patients with COVID-19 may progress to a severe fibro-proliferative phase and
ventilator dependency. ECMO in these patients would likely serve little purpose. ECMO
however may improve survival in patients with severe single organ failure (lung) if initiated
within 7 days of intubation. [235]
• Combination inhaled nitric oxide (or epoprostenol) and intravenous almitrine. The
combination of inhaled nitric oxide, a selective pulmonary vasodilator, and almitrine, a
specific pulmonary vasoconstrictor, may improve the severe V/Q mismatch in patients with
severe COVID-19 “pneumonia”. [236-239]

Page 13 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 6. Premature discontinuation of corticosteroids and IV vitamin C (after 4 day) and the effect of
reinitiation of this combination on the CRP profile.

Salvage treatments of unproven/no benefit.


• Convalescent serum/monoclonal antibodies: the role and timing of convalescent serum and
monoclonal antibodies are uncertain. [240-243] Two RCT’s failed to demonstrate any clinical
benefit with convalescent serum. [215,216] In addition, Eli Lilly recently announced that they
are suspending their ACTIV-33 clinical trial as their monoclonal antibody failed to
demonstrate a clinical benefit in hospitalized patients. It is noteworthy that the only RCT
demonstrating efficacy of convalescent plasma for an infectious disease was conducted more
than 40 years ago, for treating Argentine hemorrhagic fever. [211] Furthermore, giving
antibodies directed against SARS-CoV-2 appears pointless when the virus is already DEAD
(pulmonary phase). In addition, IgG is a large protein which penetrates tissues poorly, and is
unlikely to achieve submucosal concentrations required for mucosal immunity.[244] And
lastly, COVID-19 pulmonary disease is immune mediated, and it would therefore appear
paradoxical to enhance the antibody response with convalescent serum. [245]
• Janus Kinase inhibitors downregulate cytokine expression and may have a role in this disease.
[246-248] The role of the combination of Baricitinib and Remdesivir is unclear.[249]
• In patients with progressive fibrosis the combination of anti-fibrotic therapy with
corticosteroids should be considered. [250-253] It should however be recognized that unlike
all the medications in the MATH+ protocol, pirfenidone and nintedanib have complex side-
effects and drug interactions and should be prescribed by pulmonary physicians who have
experience with these drugs.
• CVVH/D with cytokine absorbing/filtering filters [254,255] This treatment strategy appears to
have a very limited role.

Page 14 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


25. Treatment of Macrophage Activation Syndrome (MAS)
• A sub-group of patients will develop MAS, particularly those patients with severe COVID-19
disease.[256] While the pathophysiology of MAS in the setting of COVID-19 is unclear this
appears to be driven by SARS-CoV-2 induced inflammasome activation and increased IL-18
production as well as increased GM-CSF and INFγ production. [257-260] The role of IL-1 and
IL-6 in the pathogenesis of MAS is unclear.
• A ferritin > 4400 ng/ml is considered diagnostic of MAS. Other diagnostic features include
increasing AST/ALT and CRP and progressive multi-system organ failure.[261]
• “High dose corticosteroids.” Methylprednisolone 120 mg q 6–8 hourly for at least 3 days,
then wean according to Ferritin, CRP, AST/ALT (see Figure 8). Ferritin should decrease by at
least 15% before weaning corticosteroids.
• Consider plasma exchange.
• The role of inhibition of IL-1 (Anakinra) and IFNγ (emapalumab) is unclear (NCT04324021).

26. Monitoring
• On admission: Procalcitonin (PCT), CRP, BNP, Troponins, Ferritin, Neutrophil-Lymphocyte
ratio, D-dimer and Mg. CRP and D-dimer are important prognostic markers. A PCT is
essential to rule out coexisting bacterial pneumonia.
• Daily: CRP, Ferritin, D-Dimer and PCT. CRP and Ferritin track disease severity closely (although
ferritin tends to lag behind CRP). Early high CRP levels are closely associated with the degree
of pulmonary involvement and the CT score. [262]
• In patients receiving IV vitamin C, the Accu-Chek™ POC glucose monitor will result in
spuriously high blood glucose values. Therefore, a laboratory glucose is recommended to
confirm the blood glucose levels. [263,264]
• No routine CT scans, follow CXR and chest ultrasound.
• ECHO as clinically indicated; Pts may develop a severe “septic” cardiomyopathy.

Page 15 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 7.

Page 16 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 8. SARS-CoV-2 induced Macrophage Activation Syndrome (MAS) treated with Vitamin
C 3g IV q 6 and increased methylprednisolone (125 mg q 8 hourly)

Page 17 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


27. Post ICU management

a. Enoxaparin 40–60 mg s/c daily


b. Methylprednisolone 40 mg day, then wean slowly (follow CRP)
c. Vitamin C 500 mg PO BID
d. Melatonin 3–6 mg at night
e. Vascepa, Lovaza or DHA/EPA 4g day (important for resolution of inflammation)

28. Post Hospital Discharge management

a. Patients have an increased risk of thromboembolic events post-discharge. [265] Extended


thromboprophylaxis (? with a DOAC) should be considered in high risk patients. Risk factors
include:[266]
i. Increased D dimer (> 2 times ULN)
ii. Increased CRP (> 2 times ULN) [267]
iii. Age > 60
iv. Prolonged immobilization

b. The post-COVID-19 syndrome (long haul syndrome), is characterized by prolonged malaise,


headaches, generalized fatigue, painful joints, dyspnea, chest pain and cognitive
dysfunction.[268-273] Up to 50% of patients experience prolonged illness after Covid-19. The
post-COVID-19 syndrome may persistent for months after the acute infection and almost
half of patients report reduced quality of life. The neurological symptoms may be related
micro- and/or macrovascular thrombotic disease which appears to be common in severe
COVID-19 disease.[256] Brain MRIs’ 3 months post-infection demonstrated micro-structural
changes in 55% of patients. [274] Similar to patients who have recovered from septic shock,
[275] a prolonged (many months) immune disturbance with elevated pro- and anti-
inflammatory cytokines may contribute to the post-COVID-19 syndrome. Consequently, A
CRP should be measured prior to discharge and a tapering course of corticosteroids should
be considered in those with an elevated CRP. It should be noted that much like omega-3
fatty acids corticosteroids have been demonstrated to increase expression of pro-resolving
lipids including Protectin D1 and Resolvin D4.[276] Other interventions that should be
considered include:
i. Vascepa, Lovaza or DHA/EPA 4g day; important for resolution of inflammation by
inducing resolvin production. [91,92]
ii. Atorvastatin 40 mg daily (increase resolvin synthesis) [277]
iii. Continue melatonin for its antioxidant properties and stabilization of the circadian
rhythms.
iv. Multivitamin with adequate vitamin D.
v. Recently Ivermectin has been reported to have a role in the treatment of post-
Covid-19 syndrome. [278] The anti-inflammatory properties of ivermectin may
mediate this benefit.

c. Post-COVID-19 pulmonary fibrosis. An unknown number of patients who have recovered


from COVID-19 organizing pneumonia will develop pulmonary fibrosis with associated
limitation of activity. These patients should be referred to a pulmonologist with expertise in
pulmonary fibrosis. Anti-fibrotic therapy may have a role in these patients, [250-253]
however additional data is required before this therapy can be more generally
recommended.

Page 18 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


29. Maintaining mental health and the avoiding the misinformation pandemic

‘Misinformation on the Coronavirus might be the most contagious thing about it”
Dr. Tedros, WHO Director General

• The Panic and misinformation spread by Social Media travels faster than the pandemic
itself. What you can do?
o Avoid social media as much as possible; excess social media exposure increases
the likelihood of anxiety and depression[279]
o Read the news/information from reliable sources (if you can find one)
o Have a designated time for checking information
o People share false claims about COVID-19 partly because they simply fail to
think sufficiently about whether or not the content is accurate when deciding
what to share. [280]
o Stay connected to positive people! Remotely!
o Have a plan for staying in touch with family and friends
o Identify positive influencers…limit contact with other “worriers”
o Isolation can cause rumination/anxious thinking to escalate
o Maintain a sense of hope, humanity and kindness toward others
o Seek professional help if anxiety is overwhelming
• Recognize the things you can control
o WEAR A MASK when in contact with others
o Establish social distancing; stand/sit about 6 feet away from others
o Limit attendance at large gatherings
o Eliminate your contact with those who are ill
o DON’T go to work or school if you are sick
o Practice self-care
 Good sleep, balanced diet, exercise
 Mindfulness/Meditation/Relaxation activities

Page 19 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Key Concepts of the FLCCC Alliance Treatment Protocol
This is a very complex disease; many of the mysteries are still unravelling. However, a number of
concepts are key to the management of this “treatable disease; they include.

1. Patients transition through a number of different phases (clinical stages). The treatment of each
phase is distinct ... this is critically important (see Figures 1 & 2).
2. Antiviral therapy is likely to be effective only during the viral replicative phase whereas anti-
inflammatory therapy is expected to be effective during the pulmonary phase and possibly the
post-COVID-19 phase. While Remdesivir is a non-specific antiviral agent that targets RNA
viruses, it is likely that agents specifically designed to target SARS-CoV-2 will be developed.
3. The SARS-CoV-2 PCR remains positive for at least 2 weeks following detection of whole virus (by
culture, See figure 3). Patients who progress to the pulmonary phase are usually PCR positive
despite cessation of viral replication (and are therefore less likely to be infectious).
4. Due to the imperfect sensitivity of the PCR test as many as 20% of patients who progress to the
pulmonary phase will be PCR negative (even on repeat testing). At symptom onset PCR will be
positive in approximately 60% of patients; maximal positivity rate is on day 8 (post infection)
when 80% of patients will be positive (see Figure3). [281]
5. Symptomatic patients are likely to be infectious during a narrow window starting 2–3 days
before the onset of symptoms and to up to 6 days after the onset of symptoms (see Figure
3).[282]
6. It is important to recognize that COVID-19 patients present with a variety of phenotypes, likely
dependent on inoculum size and viral load, genetic heterogeneity mutations and
polymorphisms, biotypes, blood type, sex and androgen status, age, race, BMI (obesity),
immunological and nutritional status, and co-morbidities.[157,283-293] The phenotype at
presentation determines the prognosis and impacts the optimal approach to treatment.
7. The pulmonary phase is characterized by immune dysregulation, [246,248,256,259,260,286,294-
303] a pulmonary microvascular injury (vasculopathy),[256,303-306] with activation of clotting
and a pro-coagulant state together with the characteristics of an organizing pneumonia.
[307,308]
8. Endothelial damage and an imbalance of both innate and adaptive immune responses, with
aberrant macrophage activation, plays a central role in the pathogenesis of the severe COVID-19
Disease. [256]
9. As patients, progress down the pulmonary cascade the disease becomes more difficult to
reverse. The implications of this are twofold.
a. Early treatment (of the pulmonary phase) is ESSENTIAL to a good outcome.
b. Treatment in the late pulmonary phase may require escalation of the dose of
corticosteroids as well as the use of salvage methods (i.e. plasma exchange). However,
patients who present in the late pulmonary phase may have progressed to the
irreversible pulmonary fibroproliferative phase (see Figure 9).
10. The pulmonary phase of COVID-19 is a treatable disease; it is inappropriate to limit therapy to
“supportive care” alone. Furthermore, it is unlikely that there will be a single “silver bullet” to
treat severe COVID-19 disease. Rather, patients will require treatment with multiple
drugs/interventions that have synergistic and overlapping biological effects. Repurposed FDA
approved drugs that are safe, inexpensive, and “readily” available are likely to have a major
therapeutic effect on this disease. The impact of COVID-19 on middle- and low-income countries
is enormous; these countries are not able to afford expensive propriety “designer” molecules.
11. The radiographic and pathological finding of COVID-19 lung disease are characteristic of a
secondary organizing pneumonia (and not ARDS). [307,309,310]

Page 20 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


12. THIS is NOT ARDS (at least initially). The initial pulmonary phase neither looks like, smells like
nor is ARDS.[311-313] The ground glass infiltrates are peripheral and patchy, [309] and do not
resemble the dependent air space consolidation (sponge/baby lung) seen with “typical
ARDS”.[314] Extravascular lung water index (EVLWI) is normal or only slightly increased; this by
definition excludes non-cardiogenic pulmonary edema (ARDS). Lung compliance is normal (this
excludes ARDS). Patients are PEEP unresponsive. Treating patients as if they ARDS is a very
dangerous approach. The hypoxia is due to severe ventilation/perfusion mismatch likely due to
the microvascular narrowing, thrombosis and vasoplegia.
13. The core principles of the pulmonary phase (MATH+) is the use of anti-inflammatory agents to
dampen the “cytokine storms” together with full anticoagulation to limit the microvascular and
macrovascular clotting and supplemental oxygen to help overcome the hypoxia.
14. Ivermectin has emerged as the “wonder drug” to prophylaxis and treat COVID-19. Ivermectin
inhibits viral replication and has potent anti-inflammatory properties. Emerging clinical data
(including RCT’s) suggest that ivermectin may have an important clinical benefit across the
spectrum of phases of the disease, i.e pre-exposure prophylaxis, postexposure prophylaxis,
during the symptomatic phase and during the pulmonary phase. [1-3,67,70-77,134-136,187-
193,315] In the recommended dosages, Ivermectin is remarkably safe (see Table 1 and Fig 5).
However, as noted above there is the potential for serious drug-drug interaction.
15. The pulmonary phase of COVID-19 is characterized by PROLONGED immune dysregulation that
may last weeks or even months. The early and abrupt termination of anti-inflammatory agents
will likely result in rebound inflammation (see Figure 8).[316]
16. SARS-CoV-2 as compared to all other respiratory viruses, upregulates cytokines and chemokines
while at the same time down regulating the expression of Interferon alpha (the hosts primary
antiviral defence mechanism). [131,155] Low innate antiviral defenses and high pro-
inflammatory mediators contribute to ongoing and progressive lung injury.
17. Patients in whom the cytokine storm is not “dampened” will progress into the “H phenotype”
characterized by poor lung compliance, severe oxygenation failure and PEEP recruitability (see
Figure 9). Progression to this phase is exacerbated by ventilator induced lung injury (VILI). The
histologic pattern of the “H Phenotype” is characterized by an acute fibrinous and organizing
pneumonia (AFOP), with extensive intra-alveolar fibrin deposition called fibrin “balls” with
absent or minimal hyaline membranes.[288,310,317-319] Corticosteroids seem to be of little
benefit in established AFOP. High dose methylprednisolone should be attempted in the “early
phase” of AFOP, however many patients will progress to irreversible pulmonary fibrosis with
prolonged ventilator dependency and ultimately death.
18. An unknown percentage of patients with COVID-19 present with “silent hypoxia” with a blunted
respiratory response. This phenomenon may be related to involvement of chemoreceptors of
the carotid bodies and/or brain stem dysfunction,[320,321] and necessitates pulse oximetry in
symptomatic patients managed at home (as discussed above).
19. It should be recognized that LWMH has non-anticoagulant properties that are likely beneficial in
patients with COVID-19, these include anti-inflammatory effects and inhibition of histones.[322]
in addition, in vitro studies demonstrate that heparin inhibits SARS-CoV-2 interaction with the
ACE-2 receptor and viral entry,[323,324] as well as viral replication [75,138]. Most importantly
LWWH inhibits heparanase (HPSE).[325] HSE destroys the endothelial glycocalyx increasing
endothelial leakiness, activating clotting and potentiating endothelialitis.[325] HPSE levels have
been reported to be increased in patients with severe COVID-19 infection. [326]
20. Due to the ease of administration, greater anti-Xa activity and better safety profile we prefer
low molecular weight heparin (LMWH) to unfractionated heparin (UFH).
21. The combination of steroids and ascorbic acid (vitamin C) is essential. Both have powerful
synergistic anti-inflammatory actions. [173,181] Vitamin C protects the endothelium from
oxidative injury.[174,327-329] Furthermore, vitamin C Increases the expression of interferon-

Page 21 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


alpha [26] while corticosteroids (alone) decease expression of this important protein. [330-
333] It should be noted that when corticosteroids are used in the pulmonary phase (and not in
the viral replicative phase) they do not appear to increase viral shedding or decrease the
production of type specific antibodies. [159,334] It is likely that heparin (LMWH) acts
synergistically with corticosteroids and vitamin C to protect the endothelium and treat the
endothelialitis of severe COVID-19 disease.
22. Notwithstanding the very important and impressive results of the Recovery-Dexamethasone
study, methylprednisolone is the corticosteroid of choice for the pulmonary phase of COVID-19.
This is based on pharmacokinetic data (better lung penetration),[335] genomic data specific for
SARS-CoV-2,[336] and a long track record of successful use in inflammatory lung diseases. (see
Table 1)
23. For prophylaxis and treatment of the early symptomatic phase we suggest the combination of
Quercetin (a plant polyphenol), Vitamin C and Zinc.[35] This is based on intriguing basic science
data which indicates that:
a. Zinc is essential for innate and adaptive immunity.[52] In addition, Zinc inhibits RNA
dependent RNA polymerase in vitro against SARS-CoV-2 virus.[51]
b. Quercetin has direct viricidal properties against a range of viruses, including SARS-CoV-
2, and is a potent antioxidant and anti-inflammatory agent. [24,29,34,34,337-344] In
addition, quercetin acts as a zinc ionophore. [345]
c. Vitamin C improves the potency of Quercetin and has antiviral and anti-inflammatory
activity.[24]
24. It should also be noted that Vitamin D may be an enormously powerful prophylactic strategy
against COVID-19. Vitamin D deficiency explains, in part, the enormous geographic variation in
mortality of this disease. [11,346] It is likely that vitamin D supplementation may have a greater
role in the prophylaxis than the treatment of severe disease.

Page 22 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Figure 9. The consequences of “steroid” avoidance”. CT scan after 23 days of “supportive
care” demonstrating the late fibroproliferative (irreversible) phase of COVID-19 lung disease
(Image kindly provide by Dr. Pierre Kory, from NYC).

Page 23 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


Scientific Rationale for MATH+ Treatment Protocol (pulmonary phase)
Three core pathologic processes lead to multi-organ failure and death in COVID-19:

1) Hyper-inflammation (“Cytokine storm”) – a dysregulated immune system whose cells infiltrate


and damage the lungs as well as other organs including the heart and bone marrow. It is now
widely accepted that SARS-CoV-2 causes aberrant T lymphocyte and macrophage activation
resulting in a “cytokine storm.” [246,248,259,260,286,294,296-302] In addition, post-mortem
examination has demonstrated features of the “macrophage activation syndrome”, with
hemophagocytosis and a hemophagocytic lymphohistiocytosis-like disorder.[256]
2) Hyper-coagulability (increased clotting) – the dysregulated immune system damages the
endothelium and activates blood clotting, causing the formation of micro and macro blood clots.
Clotting activation may occur directly due to increased expression of Factor Xa as well as
endothelial injury with the release of large aggregates of van Willebrand factor.[81] These blood
clots impair blood flow. [139,140,142-150,305,306,347,348] It should be noted that the
thrombotic microangiopathy appears to target predominantly the pulmonary and cerebral
circulation. [256]
3) Severe Hypoxemia (low blood oxygen levels) –lung inflammation caused by the cytokine storm,
together with microthrombosis in the pulmonary circulation severely impairs oxygen absorption
resulting in oxygenation failure.

The above pathologies are not novel, although the combined severity in COVID-19 disease is
considerable. Our long-standing and more recent experiences show consistently successful treatment if
traditional therapeutic principles of early and aggressive intervention is achieved, before the onset of
advanced organ failure. It is our collective opinion that the historically high levels of morbidity and
mortality from COVID-19 is due to a single factor: the widespread and inappropriate reluctance amongst
hospitalists and intensivists to employ anti-inflammatory and anticoagulant treatments, including
corticosteroid therapy early in the course of a patient’s hospitalization. It is essential to recognize that
it is not the virus that is killing the patient, rather it is the patient’s overactive immune system.
[245,248,256,321] Autopsy studies have demonstrated minimal viral cytopathic effects.[256,321] The
flames of the “cytokine fire” are out of control and need to be extinguished. Providing supportive care
(with ventilators that themselves stoke the fire) and waiting for the cytokine fire to burn itself out simply
does not work… this approach has FAILED and has led to the death of tens of thousands of patients.

“If what you are doing ain’t working, change what you are doing” – PEM
The systematic failure of critical care systems to adopt corticosteroid therapy (early in this pandemic)
resulted from the published recommendations against corticosteroids use by the World Health
Organization (as recent as May 27th 2020) [349,350]. This recommendation was then perpetuated by the
Centers for Disease Control and Prevention (CDC), the American Thoracic Society (ATS), Infectious
Diseases Association of America (IDSA) amongst others. A publication authored one of the members of
the Front Line COVID-19 Critical Care (FLCCC) Alliance (UM), identified the errors made by these
organizations in their analyses of corticosteroid studies based on the findings of the SARS and H1N1
pandemics.[154,351] Their erroneous recommendation to avoid corticosteroids in the treatment of
COVID-19 has led to the development of myriad organ failures which have overwhelmed critical care
systems across the world and led to excess deaths. The recently published results of the RECOVERY-
DEXAMETHASONE study provide definitive and unambiguous evidence of the lifesaving benefits of
corticosteroids and strong validation of the MATH + protocol. It should be recognized that
corticosteroids are the only therapy proven to reduce the mortality in patients with COVID-19.[352] The

Page 24 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


RECOVERY-DEXAMETHASONE study, randomized 2104 patients to receive dexamethasone 6 mg
(equivalent to 32 mg methylprednisolone) once per day (either by mouth or by intravenous injection)
for ten days and were compared with 4321 patients randomized to usual care alone.[128]
Dexamethasone reduced deaths by one-third in ventilated patients (rate ratio 0.65 [95% confidence
interval 0.48 to 0.88]; p=0.0003) and by one fifth in other patients receiving oxygen only (0.80 [0.67 to
0.96]; p=0.0021). There was no benefit among those patients who did not require respiratory support
(1.22 [0.86 to 1.75; p=0.14). The results of this study STRONGLY support the EVMS/MATH+ protocol
which recommends the use of corticosteroids for the “pulmonary phase” of COVID-19. It should be
noted that we would consider the non-titratable ‘fixed” dose of dexamethasone used in the RECOVERY-
DEXAMETHASONE study to be very low. Furthermore, as indicated above we consider
methylprednisolone to be the corticosteroid of choice for the treatment of COVID-19 pulmonary
disease. The benefit of methylprednisolone in improving respiratory function, ventilator dependency
and mortality has been confirmed in a number of observational studies, [155,156,162,334,353-355] as
well as a randomized controlled study.[164] It should be recognized that the mortality benefit with
methylprednisolone was not replicated in a recent Brazilian RCT. [316] However, in this study
methylprednisolone was started relatively late (day 13 after symptom onset), but most importantly was
stopped on day 5. This failed study reinforces the concept of early and prolonged treatment with
methylprednisolone titrated to the patient’s clinical response. In patients at high risk of Strongyloides
infection, screening and/or treatment of this parasite with ivermectin is suggested prior to treatment
with corticosteroids.[356]

Our treatment protocol targeting the key pathologic processes has been highly successful,
if begun within 6 hours of a COVID19 patient presenting with shortness of breath and/or arterial
desaturation and requiring supplemental oxygen. If such early initiation of treatment could be
systematically achieved, the need for mechanical ventilators and ICU beds will decrease dramatically.

Further resources:

The reader is referred to the large autopsy series by Bruce and colleagues which clearly outlines the
pathophysiology of severe COVID-19 disease.[256]

The scientific rationale for the MATH + protocol is reviewed in this paper.[171]

In this U-tube video, Professor Britt Glaunsinger, PhD provides an outstanding review on the molecular
virology of SARS-CoV-2: https://www.youtube.com/watch?v=DQVpHyvz4no

Lectures by Paul Marik, MD reviewing clinical aspects of COVID-19.


https://www.youtube.com/channel/UCz9Pvn15m4Rv1uY-aBYRVuw

Page 25 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


References

1. Gorial FI, Mashhadani S, Sayaly HM, Dakhil BD, AlMashhadani MM. Effectiveness of Ivermectin as add-on
therapy in COVID-19 management (Pilot Trial). medRxiv 2020.
2. Khan MS, Khan MS, Debnath Cr, Nath PN, Mahtab MA. Ivermectin treatment may improve the prognosis
of patients with COVID-19. Archivos de Bronconeumologia 2020.
3. Rajter JC, Sherman MS, Fatteh N, Vogel F, Sacks J, Rajter JJ. ICON (Ivermectin in COvid Ninteen) study: Use
of ivermectin is associated with lower mortality in hospitalized patients with COVID-19. Chest 2020.
4. Niaee MS, Gheibl N, Namdar P, Allami A, Javadi A. Ivermectin as an adjunct treatment for hospitalized
adult COVID-19 patients: A randomized multi-center clinical trial. Research Square 2020.
5. Elgazzar A, Hany B, Youssef SA, Hany B, Hafez M. Efficacy and safety of ivermectin for treatment and
prophylaxis of COVID-19 pandemic. Research Square 2020.
6. Hashim HA, Maulood MF, rasheed AM, Fatak DF, Kabah KK. Controlled randomized clinical trial on using
Ivermectin with Doxycycline for treating COVID-19 patients in Bagdad, Iraq. medRxiv 2020.
7. Maghbooli Z, Sahraian MA, Ebrahimi M, Pazoki M, Kafan S. Vitamin D sufficiency, a serum 25-
hydroxyvitamin D at least 30 ng/ml reduced risk for adverse clinical outcomes in patients with COVID-19
infection. PloS ONE 2020; 15:e0239799.
8. Grant WB, Lahore H, McDonnell SL, Baggerly CA, French Cb, Aliaono JL. Evidence that Vitamin D
supplementation could reduce risk of influenza and COVID-19 infections and deaths. Nutrients 2020;
12:988.
9. Kaufman HW, Niles JK, Kroll MH, Bi C, Holick MF. SARS-CoV-2 positivity rates associated with circulating
25-hydroxyvitamin D level. PloS ONE 2020; 15:e0239252.
10. Lau FH, Majumder R, Torabi R, Saeg F, Hoffman R, Cirillo JD. Vitamin D insufficiency is prevalent in severe
COVID-19. medRxiv 2020.
11. Marik PE, Kory P, Varon J. Does vitamin D status impact mortlality from SARS-CoV-2 infection? Medicine in
Drug Discovery 2020.
12. Rhodes JM, Subramanian S, Laird E, Kenny RA. Editorial: Low population mortality from COVID-19 in
countries south of 35 degrees North - supports vitamin D as a factor determining severity. Alimentary
Pharmacology & Therapeutics 2020; (in press).
13. Dancer RC, Parekh D, Lax S et al. Vitamin D deficiency contributes directly to the acute respiratory distress
syndrome (ARDS). Thorax 2015; 70:617-24.
14. LLie PC, Stefanescu S, Smith L. The role of vitamin D in the prevention of coronavirus disease 2019
infection and mortality. Aging Clin Exp Res 2020.
15. Daneshkhah A, Eshein A, Subramanian H. The role of vitamin D in suppressing cytokine storm of COVID-19
patients and associated mortality. medRxiv 2020.
16. Bergman P, Lindh AU, Bjorkhem-Bergman L, Lindhagen L. Vitamin D and respiartory tract infections: A
systematic review and meta-analysis of randomized controlled trials. PloS ONE 2013; 8:e65835.
17. Carpagnano GE, Lecce V, Quaranta VN, Zito A, Buonamico E. Vitamin D deficiency as a predictor of poor
prognosis in patients with acute respiratory fialure due to COVID-19. J Endocrinol Invest 2020.
18. Israel A, Cicurel A, Feldhamer I et al. The link between vitamin D deficiency and Covid-19 in a large
population. medRxiv 2020.
19. Radujkovic A, Hippchen T, Tiwari-Heckler S, Dreher S, Merle U. Vitamin D deficiency and outcome of
COVID-19 patients. Nutrients 2020; 12:2757.
20. Rizzoli R. Vitamin D supplementation: upper limit for safety revisited. Aging Clin Exp Res 2020.
21. Annweiler C, Hanotte B, de L'Eprevier CG, Sabatier JM, Lafaie L. Vitamin D and survival in COVID-19
patients: A quasi-experimental study. Journal of Steroid Biochemistry & Molecular Biology 2020.
22. Moozhipurath RK, Kraft L, Skiera B. Evidence of protective role of Ultraviolet-B (UVB) radiation in reducing
COVID-19 deaths. Nature Research 2020; 10:17705.
23. Maggini S, Beveridge S, suter M. A combination of high-dose vitamin C plus zinc for the common cold.
Journal of International Medical Research 2012; 40:28-42.
24. Colunga Biancatelli RM, Berrill M, Catravas JD, Marik PE. Quercetin and Vitamin C: experimental therapy
for the prevention and treatment of SARS-CoV-2 via synergistic action. Front Immunol 2020.
25. Kyung Kim T, Lim HR, Byun JS. Vitamin C supplementaion reduces the odds of developing a common cold
in Republic of Korea Army recruits: a randomised controlled trial. BMJ Mil Health 2020.

Page 26 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


26. Colunga Biancatelli RM, Berrill M, Marik PE. The antiviral properties of vitamin C. Expert Rev Anti Infect
Ther 2020; 18:99-101.
27. Hiedra R, Lo KB, Elbashabsheh M, Gul F, Wright RM. The use of IV vitamin C for patients with COVID-19: a
case series. Exp Rev Anti Infect Ther 2020.
28. Khaerunnisa S. Potential inhibitor of COVID-19 main protease (Mpro) from several medicinal plant
compuns by molecular docking study. medRxiv 2020.
29. Chen L, Li J, Luo C, Liu H, Xu W, Chen G. Binding interaction of quercetin-3-B-galactoside and its synthetic
derivatives with SARS-CoV 3CL: structure-activity relationship reveal salient pharmacophore features.
Bioorganic & Medicinal Chemistry Letters 2006; 14:8295-306.
30. Nain Z, Rana HK, Lio P, Islam SM, Summers MA, Moni MA. Pathogenic profiling of COVID-19 and SARS-like
viruses. Briefings in Bioinformatics 2020.
31. Yi L, Li Z, Yuan K et al. Small molecules blocking the entry of severe respiratory syndrome coronavirus into
host cells. J Virol 2020; 78:11334-9.
32. Shakoor H, Feehan J, Dhaheri AS, Ali HI, Platat C, Ismail LC. Immune-boosting role of vitamins D,C,E, zinc,
selenium and omega-3 fatty acids: could they help against COVID-19. Maturitas 2020.
33. Calder PC. Nutrition, immunity and COVID-19. BMJ Nutrition, Prevenion & Health 2020; 3.
34. Abian O, Ortega-Alarcon D, Jimenez-Alesanco A, Ceballos-Laita L, Vega S. Structural stability of SARS-CoV-
2 3CLpro and identification of quercetin as an inhibitor by experimental screening. International Journal of
Biological Macromolecules 2020; 164:1693-703.
35. Arslan B, Ergun NU, Topuz S, Semerci SY, Suner N. Synergistic effect of quercetin and vitamin C against
COVID-19: Is a possible guard for front liners? ssrn 2020.
36. Giuliani C, Bucci I, Di Santo S, Rossi C, Grassadonia A, Piantelli M. The flavonoid quercetin in hibits thyroid-
restricted genes expression and thyroid function. Food and Chemical Toxicology 2014; 66:23-9.
37. de Souza dos Santos MC, Goncalves CF, Vaisman M, Ferreira AC, de Carvalho DP. Impact of flavonoids on
thyroid function. Food and Chemical Toxicology 2011; 49:2495-502.
38. Chandra AK, De N. Catechin induced modulation in the activities of thyroid hormone synthesizing
enzymes leading to hypothyroidism. Mol Cell Biochem 2013; 374:37-48.
39. Pistollato F, Masias M, Agudo P, Giampieri F. Effects of phytochemicals on thyroid function and their
possible role in thyroid disease. Ann N Y Acad Sci 2019; 1433:3-9.
40. Sathyapalan T, Manuchehri AM, Thatcher NJ, Rigby AS, Chapman T. The effect of soy phytoestrogen
supplementation on thyroid status and cardiovascular risk markers in patients with subclinical
hypothyroidism: A randomized, double-blind, crossover study. J Clin Endocrinol Metab 2020; 96:1422-49.
41. Tonstad S, Jaceldo-Siegl K, Messina M, Haddad E. The association between soya consumption and serum
thyroid-stimulating hormone in the Adventist Health Study-2. Public Health Nutr 2016; 19:1464-70.
42. Colombo D, Lunardon L, Bellia G. Cyclosporine and herbal supplement interactions. Journal of Toxicology
2014; 2014:145325.
43. Colunga Biancatelli RM, Berrill M, Mohammed YH, Marik PE. Melatonin for the treatment of sepsis: the
scientific rationale. J Thorac Dis 2020; 12 (Suppl 1):S54-S65.
44. Reiter RJ, Abreu-Gonzalez P, Marik PE, Dominguez-Rodriguez A. Therapeutic algorithm for use of
melatonin in patients with COVID-19. Front Med 2020; 7:226.
45. Reiter RJ, Sharma R, Ma Q, Dominquez-Rodriguez A, Marik PE, Abreu-Gonzalez P. Melatonin inhibits
COVID-19-induced cytokine storm by reversing aerobic glycolysis in immune cells: A mechanistic analysis.
Medicine in Drug Discovery 2020; 6:100044.
46. Zhang R, Wang X, Ni L, Di X, Ma B. COVID-19: Melatonin as a potential adjuvant treatment. Life Sci 2020;
250:117583.
47. Jehi L, Ji X, Milinovich A, erzurum S, Rubin B, Gordon S. Individualizing risk prediction for positive COVID-
19 testing. Results from 11,672 patients. Chest 2020.
48. Kleszczynski K, Slominski AT, Steinbrink K, Reiter RJ. Clinical trials for use of melatonin to fight COVID-19
are urgently needed. Nutrients 2020; 12.
49. Coto-Montes A, Boga JA. ER stress and autophagy induced by SARS-CoV-2: The targer for melatonin
treatment. Melatonin Res 2020; 3:346-61.
50. Gandolfi JV, Di Bernardo AP, Chanes DA et al. The effects of melatonin supplementation on sleep quality
and assessment of the serum melatonin in ICU patients: A randomized controlled trial. Crit Care Med
2020.

Page 27 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


51. te Velthuis AJ, van den Worm SH, Sims AC, Baric RS, Snijder EJ, van Hemert MJ. Zn2+ inhibits Coronavirus
and Arterivirus RNA polymerase activity In Vitro and Zinc ionophores block the replication of these viruses
in cell culture. PLos Pathog 2010; 6:e1001176.
52. Gammoh NZ, Rink L. Zinc in Infection and Inflammation. Nutrients 2017; 9.
53. Hemila H. Zinc lozenges and the common cold: a meta-analysis comparing zinc acetate and zinc gluconate,
and the role of zinc dosage. J Royal Soc Med Open 2017; 8:1-7.
54. Singh M, Das RR. Zinc for the common cold. Cochrane Database of Syst Rev 2013; 6:CD001364.
55. Hoeger J, Simon TP, Beeker T, Marx G, Haase H. Persistent low serum zinc is associated with recurrent
sepsis in critically ill patients - A pilot study. PloS ONE 2017; 12:e0176069.
56. Shakoor H, Freehan J, Mikkelsen K, Al Dhaheri AS, Ali HI. Be well: A potential role for vitamin B in COVID-
19. Maturitas 2020.
57. dos Santos LM. Can vitamin B12 be an adjuvant to COVID-19 treatment? GSC Biological and
Pharmaceutical Sciences 2020; 11.
58. Kandeel M, Al-Nazawi M. Virtual screening and repurposing of FDA approved drugs against COVID-19
main protease. Life Sci 2020; 251:117627.
59. Tan CW, Ho LP, Kalimuddin S, Cherng BP, Teh YE. Cohort study to evaluate effect of vitamin D,
magnesium, and vitamin b12 in combination on severe outcome progression in older patients with
coronavirus (COVID-19). Nutrition 2020; 80:111017.
60. Zhang P, Tsuchiya K, Kinoshita T, Kushiyama H, Suidasari S, Hatakeyama M. Vitamin B6 prevents IL-1B
protein production by inhibiting NLRP3 inflammasome activation. J Biol Chem 2020; 291:24517-27.
61. Behera P, Patro BK, Singh AK, Chandanshive PD, Kumar R. Role of ivermectin in the prevention of COVID-
19 infection among healthcare workers in India: A matched case-control study. medRxiv 2020.
62. Carvallo H, Hirsch RR, Alkis P, Contreras V. Study of the efficacy and safety of topical ivermectin + Iota-
carrageenan in the prophylaxis against COVID-19 in health personnel. Journal of Biomedical Research and
Clinical Investigation 2020; 2.
63. Kory P, Meduri GU, Iglesias J et al. Review of the emerging evidencce supporting the use of Ivermectin in
the prophylaxis and treatment of COVID-19. Front Line Covid-19 Critical Care Alliance. osf io 2020.
64. Hellwig MD, Maia A. A COVID-19 prophylaxis? Lower incidence associated with prophylactic administraion
of ivermectin. Int J Antimicrob Agents 2020.
65. Munoz J, Ballester MR, Antonijoan RM, Gich I, Coili E. Safety and pharmacokinetic profile of fixed-dose
ivermectin with an innovative 18mg tablet in healthy adult volunteers. PLoS Neglected Tropical Diseases
2018; 12:e0006020.
66. Castillo ME, Costa LM, Barrios JM et al. Effect of calcifediol treatment and best available therapy versus
best available therapy on intensive care unit admission and mortality among patients hospitalized for
COVID-19: a pilot randomized clinical study. J Steroid Biochem Mol Biol 2020.
67. Hashim HA, Maulood MF, rasheed AM, Fatak DF, Kabah KK. Controlled randomized clinical triaal on using
Ivermectin with Doxycycline for treating COVID-19 patients in Bagdad, Iraq. medRxiv 2020.
68. Alam MT, Murshed R, Bhiuyan E, Saber S, Alam RF, Robin RC. A case series of 100 COVID-19 positive
patients treated with combination of Ivermectin and Doxycycline. Bangladesh Coll Phys Surg 2020; 38:10-
5.
69. Chowdhury AT, Shahabz M, Karim MR, Islam J, Guo D, He D. A randomized trial of ivermectin-doxycycline
and hydrochloroquine-azithromycin therapy on COVID-19 patients. Research Square 2020.
70. Chamie J. Real-World evidence: The case of Peru, casuality between Ivermectin and COVID-19 infection
fatality rate. ResearchGate 2020.
71. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug Ivermectin inhibits the
replication of SARS-CoV-2 in vitro. Antiviral Res 2020.
72. Lehrer S, Rheinstein PH. Ivermectin docks to the SARS-CoV-2 spike receptor-binding domain attached to
ACE2. In Vivo 2020; 34:3023-6.
73. Maurya DK. A combination of Ivermectin and Doxycycline possibly blocks the viral entry and modulate the
innate immune response in COVID-19 patients. ChemRxiv 2020.
74. Yang SN, Atkinson SC, Wang C, Lee A. The broad spectrum antiviral ivermectin targets the host nuclear
transport importin alpha/beta1 heterodimer. Antiviral Res 2020; 177:104760.
75. Dayer MR. Coronavirus (2019-nCoV) deactivation via spike glycoprotein shielding by old drugs,
bioinformatic study. Preprints 2020.

Page 28 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


76. Swargiary A. Ivermectin as a promising RNA-dependent RNA polymerase inhibitor and a therapeutic drug
against SARS-CoV2: Evidence from silico studies. Research Square 2020.
77. Kircik LH, Del Rosso JQ, Layton AM, schauber J. Over 25 years of clinical experience with Ivermectin: An
overview of safety for an increasing number of indications. J Drugs Dermatol 2016; 15:325-32.
78. Kalfas S, Visvanathan K, Chan K, Drago J. The therapeutic potential of ivermectin for COVID-19: A
systematic review of mechanisms and evidence. medRxiv 2020.
79. Bianconi V, Violi F, Fallarino F, Pignatelli P, Sahebkar A, Pirro M. Is acetylsalicylic acid a safe and potentially
useful choice for adult patients with COVID-19? Drugs 2020.
80. Muller C, Karl N, Ziebuhr J, Pleschka S. D,L-lysine acetylsalicylate + glycine impairs coronavirus replication.
J Antivir Antiretovir 2020.
81. Varatharajah N. COVID-19 CLOT: What is it? Why in the lungs? Extracellular histone, "auto-activation" of
prothrombin, emperipolesis, megakaryocytes, "self-association" of Von Willebrand factor and beyond.
Preprints 2020.
82. Freedberg DE, Conigliaro J, Sobieszczyk ME, Markowitz DD. Famotidine use is associated with impoved
clinical outcomes in hospitalized COVID-19 patients: A propensity score matched retrospective cohort
study. medRxiv 2020.
83. Janowitz T, Baglenz E, Pattinson D, Wang TC, Conigliaro J. Famotidine use and quantitative symptom
tracking for COVID-19 in non-hospitalized patients: a case series. Gut 2020; 69:1592-7.
84. Mather JF, Seip RL, McKay RG. Impact of famotidine use on clinical outcomes of hospitalized COVID-19
patients. Am J Gastroenterol 2020.
85. Malone RW, Tisdall P, Fremont-Smith P, Liu Y, Huang XP, White KM. COVID-19: Famotidine, Histamine,
Mast Cells, and mechanisms. Research Square 2020.
86. Sethia R, Prasad M, Mahapatra SJ, Nischal N, Soneja M. Efficacy of famotidine for COVID-19: A systematic
review and meta-analysis. medRxiv 2020.
87. Shoaibi A, Fortin S, Weinstein R, Berlin JA. Comparative effectiveness of famotidine in hospitalized COVID-
19 patients. medRxiv 2020.
88. Yeramaneni S, Doshi P, Sands K, Cooper M, Kurbegov D, Fromell G. Famotidine use is not associated with
30-day mortality: A coarsened exact match study in 7158 hospitalized COVID-19 patients from a large
healthcare system. medRxiv 2020.
89. Hammock BD, Wang W, Gilligan MM, Panigrahy D. Eicosanoids. The overlooked storm in Coronavirus
Disease 2019 (COVID-19)? Am J Pathol 2020.
90. Das UN. Can bioactive lipids inactivate coronavirus (COVID-19)? Arch Med Res 2020; 51:282-6.
91. Lee CR, Zeldin DC. Resolvin infectious inflammation by targeting the host response. N Engl J Med 2015;
373:2183-5.
92. Serhan CN. Novel pro-resolving lipid mediators in inflammation are leads for resolution physiology.
Nature 2014; 510:92-101.
93. Idelsis Esquivel-Moynelo I, Perez-Escribano J, Duncan-Roberts Y, Dania Vazquez-Blonquist D. Effect of
combination of interferon alpha-2b and interferon-gamma or interferon alpha 2b alone for elimination of
SARS-CoV-2 viral RNA. Preliminary results of a randomized controlled clinical trial. medRxiv 2020.
94. Davoudi-Monfarad E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M. Efficacy and safety of interferon
B-1a in treatment of severe COVID-19: A randomized clinical trial. medRxiv 2020.
95. Wang N, Zhan Y, Zhu L, Hou Z, Liu F, Song P. Retrospective multicenter cohort study shows early
interferon therapy is associated with favorable clinical responses in COVID-19 patients. Cell Host &
Microbe 2020;ePub.
96. Meng Z, Wang T, Chen L, Chen X, Li L. An experimental trial of recombinant human interferon alpha nasal
drops to prevent COVID-19 in medical staff in an epidemic area. medRxiv 2020.
97. Berg K, Bolt G, Andersen H, Owen TC. Zinc potentiates the antiviral action of human IFN-alpha tenfold. J
Interferon Cytokine Res 2001; 21:471-4.
98. Cakman I, Kirchner H, Rink L. Zinc supplementation reconstitutes the production of interferon-alpha by
leukocytes from elderly persons. J Interferon Cytokine Res 1997; 17:469-72.
99. Luks AM, Swenson ER. Pulse oximetry for monitoring patients with COVID-19 at home: Potential pitfalls
and practical guidance. Ann Thorac Med 2020.
100. Jouffroy R, Jost D, Prunet B. Prehospital pulse oximetry: a red flag for early detection of silent hypoxemia
in COVID-19 patients. Crit Care 2020; 24:313.

Page 29 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


101. Risch HA. Early outpatient treatment of symptomatic, High-Risk Covid-19 patients that should be ramped-
up immediately as key to the pandemic crisis. Am J Epidemiol 2020.
102. Borba MG, Val FF, Sampaio S. Effect of High vs Low Doses of chloroquine diphosphate as adjunctive
therapy for patietns hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
infection. A randomized clinical trial. JAMA Network Open 2020.
103. Boulware DR, Pullen MF, Bangdiwala AS, Pastick KA. A randomized trial of hydroxychloroquine as
postexposure prophylaxis for Covid-19. N Engl J Med 2020.
104. Barnabas RV, Brown ER, Bershteyn A, Johnston C, Thorpe LE. Hydroxychloroquine as postexposure
prophylaxis to prevent severe acute respiratory syndrome coronavirus 2 infection. Ann Intern Med 2020.
105. Mitja O, Corbacho-Monne M, Ubals M, Tebe C, Penafiel J. Hydroxychloroquine for early treatment of
adults with mild Covid-19: A randomized-controlled trial. Clin Infect Dis 2020.
106. Mitja O, Ubals M, Corbach-Monne M, Alemany A, Suner C. A cluster-randomized trial of
hydroxychloroquine as prevention of Covid-19 transmission and disease. N Engl J Med 2020.
107. Cavalcanti AB, Zampieri FG, Rosa RG, Azevedo LC, Veiga VC, Avezum A. Hydroxychloroquine with or
without azithromycin in mild-to-moderate Covid-19. N Engl J Med 2020; 383:2041-52.
108. Skipper CP, Pastick KA, Engen NW. Hydroxychlooquine in nonhospitalized adults with early COVID-19. Ann
Intern Med 2020; 173:623-31.
109. Rosenberg ES, Dufort EM, Udo T, Wilberschied LA. Association of treatment with hydroxychloroquine or
azithromycin with in-hospital mortality in patients with COVID-19 in New York State. JAMA 2020;
323:2493-502.
110. Geleris J, Sun Y, Platt J, Zucker J. Observational study of hydroxychloroquine in hospitalized patients with
Covid-19. N Engl J Med 2020.
111. Magagnoli J, Narendran S, Pereira F. Outcomes of hydroxychloroquine usage in United states veterans
hospitalized with COVID-19. medRxiv 2020.
112. Lopez A, Duclos G, Pastene B, Bezulier K, Guihaumou R, Solas C. Effects of hydroxychloroquine on Covid-
19 in Intensive Care Unit Patients: Preliminary Results. Int J Antimicrob Agents 2020.
113. Mahevas M, Tran VT, Roumier M, Chabrol A, Paule R. No evidence of clinical efficacy of
hydroxychloroquine in patients hospitalized for COVID-19 infection and requiring oxygen: results of a
study using routinely collected data to emulate a target trial. medRxiv 2020.
114. Elsawah HK, Elsokary MA, Elrazzaz MG, ElShafey MG. Hydroxychloroquine for treatment of non-severe
COVID-19 patients: systematic review and meta-analysis of controlled clinical trials. medRxiv 2020.
115. Axfors C, Schmitt AM, Janiaud P, van 't Hooft J, Abd_elsalam S, Abdo EF. Mortality outcomes with
hydroxychloroquine and chloroquine in COVID-19: an international collaborative meta-analysis of
randomized trials. medRxiv 2020.
116. Sbidian E, Josse J, Lemaitre G et al. Hydroxychloroquine with or without azithromycin and in-hospital
mortality or discharge in patients hospitalized for COVID-19 infection: a cohort study of 4,642 in-patients
in France. medrx 2020.
117. Effect of hydroxychloroquine in hospitalized patients with COVID-19. N Engl J Med 2020; 383:2030-40.
118. Abd-Elsalam S, Esmail ES, Khalaf M, Abdo EF, Medhat MA. Hydroxychloroquine in the Treatment of
COVID-19: A Multicenter Randomized Controlled Study. Am J Trop Med Hyg 2020; 103:1635-9.
119. Rajasingham R, Bangdiwala AS, Nicol MR, Skipper CP, Pastick KA. Hydroxychloroquine as pre-exposure
prophylaxis for COVID-19 in healthcare workers: a randomized trial. medRxiv 2020.
120. Self WE, Semler MW, Leither Lm et al. Effect of hydroxychloroquine on clinical status at 14 days in
hospitalized patients with COVID-19. a randomized clinical trial. JAMA 2020.
121. Tett SE, Cutler DJ, Day RO, Brown KF. Bioavailability of hydroxychloroquine tablets in healthy volunteers.
Br J Clin Pharmac 1989; 27:771-9.
122. MacGowan A, Hamilton F, Bayliss M, Read L, Attwood M. Hydroxychloroquine serum concentrations in
non-critical care patients infected with SARS-CoV-2. medRxiv 2020.
123. Nicol MR, Joshi A, Rizk ML, Sabato PE, Savic RM. Pharmacokinetic and pharmacological properties of
chloroquine and hydroxychloroquine in the context of COVID-19 infection. medRxiv 2020.
124. Gautret P, Lagier JC, Parola P, Hoang VT. Hydroxychloroquine and azithromycin as a treatment of COVID-
19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents 2020.
125. Lagier JC, Million M, Gautret P, Colson P, Cortaredona S, Raoult D. Outcomes of 3,737 COVID-19 patients
treated with hydroxychloroquine/azithromycin and other regimens in Marseille, France: a retrospective
analysis. Travel Medicine and Infectious Disease 2020.

Page 30 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


126. Million M, Gautret P, Colson P, Roussel Y, Dubourg G, Raoult D. Clinical efficacy of chloroquine derivatives
in COVID-19 infection: Comparative meta-analysis between big data and the real world. New Microbes
and New Infections 2020.
127. Morgan A, Stevens J. Does Bacopa monnieri improve memoy performance in older persons? Results of a
randomized, placebo-controlled, double-blind trial. J Altern Complement Med 2010; 16:753-9.
128. Effect of Dexamethasone in hospitalized patients with COVID-19-Preliminary report. N Engl J Med 2020.
129. Schultze A, Walker AJ, MacKenna B, Morten CE, Bhaskaran K, Brown JP. Inhaled corticosteroids use and
the risk of COVID-19 related death among 966,461 patients with COPD or asthma: An OpenSAFELY
analysis. medRxiv 2020.
130. Quesada-Gomez JJ, Bouillon R. Is calcifediol better than cholecalciferol for vitamin D supplementation?
Osteoporosis International 2018; 29:1697-711.
131. Cesareo R, Falchetti A, Attanasio R, Tabacco G, Naciu AM. Hypovitaminosis D: Is it time to consider the use
of calcifediol? Nutrients 2019; 11:1016.
132. Early high-dose vitamin D3 for critically ill, vitamin D-deficient patients. N Engl J Med 2019; 381:2529-40.
133. Amrein K, Martucci G, McNAlly JD. When not to use meta-analysis: Analysing the meta-analysis on vitamin
D in critical care. Clin Nutr 2017; 36:1729-30.
134. Zhang X, Song Y, Ci X, An N, Ju Y. Ivermectin inhibits LPS-induced production of inflammatory cytokines
and improves LPS-induced survival in mice. Inflamm Res 2008; 57:524-9.
135. Ci X, Li H, Yu Q et al. Avermectin exerts anti-inflammatory effect by downregulating the nuclear
transcription factor kappa-B and mitogen activated protein kinase pathway. Fundamental & Clinical
Pharmacology 2009; 23:449-55.
136. DiNicolantonio JJ, Barroso-Arranda J, McCarty M. Ivermectin may be a clinically useful anti-inflammatory
agent for late-stage COVID-19. Open Heart 2020; 7:e001350.
137. Hsu A, Liu Y, Zayac AS, Olszewski AJ, Reagan JL. Intensity of anticoagulation and survival in patients
hospitalized with COVID-19 pneumonia. Thrombosis Research 2020.
138. Kwon PS, Oh H, Kwon SJ et al. Sulphated polysaccharides effectively inhibit SARS-CoV-2 in vitro. Cell
Discovery 2020; 6:50.
139. Bikdeli B, Madhavan MV, Jimenez, Chuich T, Dreyfus I. COVID-19 and thrombotic or thromboembolic
disease: Implications for prevention, antithrombotic therapy, and follow-up. J Am Coll Cardiol 2020.
140. Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020.
141. Nadkarni GN, Lala A, Bagiella E, Chang HL, Moreno P. Anticoagulation, mortality, bleeding and pathology
among patients hospitalized with COVID-19: A single Health System Study. J Am Coll Cardiol 2020.
142. Klok FA, Kruip MJ, van der Meer NJ, Arbous MS. Incidence of thrombotic complications in critically ill ICU
patients with COVID-19. Thrombosis Research 2020.
143. Zhai Z, Li C, Chen Y, Gerotziafas G, Zhang Z. Prevention and treatment of venous thromboembolism
assocaited with Coronavirus Disease 2019 Infection: A consensus statement before guidelines. Thromb
Haemost 2020.
144. Paranjpe I, Fuster V, Lala A, Russak A, Glicksberg BS. Association of treatment dose anticoagulation with
in-hospital survival among hospitalized patietns with COVID-19. J Am Coll Cardiol 2020.
145. Iba T, Levy JH, Levi M, Connors JM. Coagulopathy of coronavirus disease 2019. Crit Care Med 2020.
146. Joly BS, Siguret V, Veyradier A. Understanding pathophysiology of hemostasis disorders in critically ill
patients with COVID-19. Intensive Care Med 2020; 46:1603-6.
147. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M. High risk of thrombosis in patients with
severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020; 46:1089-
98.
148. Varatharajah N, Rajah S. Microthrombotic complications of COVID-19 are likely due to embolism of
circulating endothelial derived ultralarge Von Willebrand Factor (eULVWF) decorated-platelet strings.
Federal Practitioner 2020.
149. Du L, Kao RY, Zhou Y, He Y. Cleavage of spike protein of SARS coronavirus by protease factor Xa is
associated with viral infectivity. Biochemical & Biophysical Research Communications 2007; 359:174-9.
150. Taccone FS, Gevenois PA, Peluso L, Pletchette Z, Lheureux O. Higher intensity thromboprophylaxis
regimens and pulmonary embolism in critically ill coronavirus disease 2019 patients. Crit Care Med 2020.
151. Hottz ED, Azevedo-Quintanilha Ig, Palhinha L, Teixeira L, Barreto EA. Platelet activation and platelet-
monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood
2020; 136:1330-41.

Page 31 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


152. Barrett TJ, Lee AH, Xia Y, Lin LH, Black M, Cotzia P. Platelet and vascular biomarkers associate with
thrombosis and death in coronavirus disease. Circulation Research 2020; 127:945-7.
153. Zhang S, Liu Y, Wang X, Yang L, Li H, Wang Y. SARS-CoV-2 binds platelet ACE2 to enhance thrombosis in
COVID-19. Journal of hematology & oncology 2020; 13:120.
154. Villar J, Confalonieri M, Pastores SM, Meduri GU. Rationale for prolonged corticosteroid tratment in the
acute respiratory distress syndrome (ARDS) caused by COVID-19. Crit Care Expl 2020; 2:e0111.
155. Fadel R, Morrison AR, Vahia A et al. Early course corticosteroids in hospitalized patients with COVID-19.
Clin Infect Dis 2020; 71:2114-20.
156. Chroboczek T, Lacoste M, Wackenheim C, Challan-Belval T, Amar B, Boisson T. Beneficial effect of
corticosteroids in severe COVID-19 pneumonia: a propensity score matching analysis. medRxiv 2020.
157. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S. Risk factors associated with acute respiratory distress syndrome
and death in patients with Coronavirus disease 2019 pneumonia in Wuhan,China. JAMA Intern Med 2020.
158. Cruz AF, Ruiz-Antoran B, Gomez AM, Lopez AS. Impact of glucocorticoid treatment in SARS-CoV-2
infection mortality: A retrospective controlled cohort study. medRxiv 2020.
159. Liu J, Zheng X, Huang Y, Shan H, Huang J. Successful use of methylprednisolone for treating severe COVID-
19. J Allergy Clin Immunol 2020.
160. Meduri GU, Bridges L, Shih MC, Marik PE, Siemienluk RA, Kocak M. Prolonged glucocorticoid treatment is
associated with improved ARDS outomces: analysis of individual patients' data from four randomized
trials and trial-level meta-analysis of the updated literature. Intensive Care Med 2016; 42:829-40.
161. Association between administration of systemic corticosteroids and mortality among critically ill patients
with COVID-19. A meta-analysis. JAMA 2020.
162. Ruiz-Irastorza G, Pijoan JI, Bereciatua E, Dunder S, Dominguez J, Garcia-Escudero P. Second week methyl-
prednisolone pulses improve prognosis in patients with severe coronavirus disease 2019 pneumonia: An
observational comparative study using routine care data. medRxiv 2020.
163. Tomazini BM, Maia IS, Cavalcanti AB, Berwanger O, Rosa RG, Veiga VC. Effect of dexamethasone on days
alive and ventilaor-free in patients with moderate or severe acute respiratory distress syndrome and
COVID-19. The CoDEX randomized clinical trial. JAMA 2020.
164. Edalatifard M, Akhtari M, Salehi M, Naderi Z, Jamshidi A, Mostafaei S. Intravenous methylprednisolone
pulse as a treatment for hospitalized severe COVID-19 patients: results from a randomised controlled
clinical trial. Eur Respir J 2020.
165. Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19. The REMAP-
CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA 2020.
166. Dequin PF, Heming N, Meziani F, Plantefeve G, Voiriot G. Effect of hydrocortisone on 21-day mortality or
respiratory support among critically ill patients with COVID-19. A randomized Clinical trial. JAMA 2020.
167. Wang Y, Zhang D, Du G, Du R. Remdesivir in adults with severe COVID-19: a randomised, double-blind,
placebo-controlled, multicenter trial. Lancet 2020; 395:1569-78.
168. Beigel JH, Tomashek KM, Dodd LE et al. Remdesivir for the treatment of Covid-19-Preliminary report. N
Engl J Med 2020;ePub.
169. Spinner CD, Gottlieb RL, Criner GJ, Lopez JR, Cattelan AM. Effect of remdesivir vs standard care on clinical
status at 11 days in patients with moderate COVID-19. A randomized clinical trial. JAMA 2020.
170. Pan H, Peto R, Karim QA, Alejandria M, Henao-Restrepo AM. Repurposed antiviral drugs for COVID-19 -
interim WHO SOLIDARITY trial. medrx 2020.
171. Marik PE, Kory P, Varon J, Iglesias J, Meduri GU. MATH+ protocol for the treatment of SARS-CoV-2
infection: the scientific rationale. Exp Rev Anti Infect Ther 2020.
172. Marik PE, Khangoora V, Rivera R, Hooper MH, Catravas J. Hydrocortisone, Vitamin C and Thiamine for the
treatment of severe sepsis and septic shock: A retrospective before-after study. Chest 2017; 151:1229-38.
173. Barabutis N, Khangoora V, Marik PE, Catravas JD. Hydrocortisone and Ascorbic Acid synergistically protect
and repair lipopolysaccharide-induced pulmonary endothelial barrier dysfunction. Chest 2017; 152:954-
62.
174. Marik PE. Hydrocortisone, Ascorbic Acid and Thiamine (HAT therapy) for the treatment of sepsis. Focus on
ascorbic acid. Nutrients 2018; 10:1762.
175. Marik PE. Vitamin C for the treatment of sepsis: The scientific rationale. Pharmacol Therapeut 2018;
189:63-70.
176. Cheng RZ. Can early and high-dose vitamin C prevent and treat coronavirus disease 2019 (COVID-19).
Medicine in Drug Discovery 2020.

Page 32 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


177. Wang Y, Lin H, Lin BW, Lin JD. Effects of different ascorbic acid doses on the mortality of critically ill
patients: a meta-analysis. Ann Intensive Care 2019; 9:58.
178. Fowler AA, Truwit JD, Hite D et al. Vitamin C Infusion for TReatment In Sepsis-Induced Acute Lung Injury-
CITRIS-ALI: A Randomized, Placebo Controlled Clinical Trial. JAMA 2018; 322:1261-70.
179. Boretti A, Banik BK. Intravenous vitamin C for reduction of cytokines storm in acute respiratory distress
syndrome. PharmaNutrition 2020; 12:100190.
180. Iglesias J, Vassallo AV, Patel V, Sullivan JB, Cavanaugh J, Elbaga Y. Outcomes of metabolic resuscitation
using ascorbic acid, thiamine, and glucocorticoids in the early treatment of sepsis. Chest 2020; 158:164-
73.
181. de Melo AF, Homem-de-Mello M. High-dose intravenous vitamin C may help in cytokine storm in severe
SARS-CoV-2 infection. Crit Care 2020; 24:500.
182. Zhang J, Rao X, Li Y et al. High-dose vitamin C infusion for the treatment of critically ill COVID-19. Research
Square 2020.
183. Kumari P, Dembra S, Dembra P, Bhawna F, Gul A, Ali B. The role of vitamin C as adjuvant therapy in
COVID-19. Cureus 2020; 12:e11779.
184. Lankadeva YR, Peiris RM, Okazaki N et al. Reversal of the pathophysiological responses to Gram-negative
sepsis by megadose Vitamin C. Crit Care Med 2020.
185. Jonmarker S, Hollenberg J, Dahlberg M, Stackelberg O. Dosing of thromboprophylaxis and mortality in
critically ill COVID-19 patients. medRxiv 2020.
186. Tomasa-Irriguible TM, Martinez-Vega S, Mor-Marco E, Herraiz-Ruiz A, Raguer-Pardo L. Low molecular
weight heparins in COVID-19 patients: beware of augmented renal clearance! Crit Care 2020; 24:325.
187. Murshed MR, Bhiuyan E, Saber S, Alam RF, Robin RF. A case series of 100 COVID-19 positive patients
treated with combination of Ivermectin and Doxycycline. Bangladesh Coll Phys Surg 2020; 38:10-5.
188. Jans DA, Wagstaff KM. Ivermectin as a broad-spectrum host directed anti-viral: The real deal. Cells 2020;
9:2100.
189. Sharun K, Dhama K, Patel SK, Pathak M, Tiwari R. Ivermectin, a new candidate therapeutic against SARS-
CoV-2/COVID-19. Ann Clin Microbiol Antimicrob 2020; 19:23.
190. Peralta EG, Fimia-Duarte R, Cardenas JW, Dominguez DV, Segura RB. Ivermectin, a drug to be considered
for the prevention and treatment of SARS-CoV-2. Brief literature review. EC Veterinary Science 2020;
5:25-9.
191. Al-Jassim KB, Jawad AA, Al-Masoudi EA, Majeed SK. Histopathological and biochemical effects of
ivermectin on kidney functions, lung and the ameliorative effects of vitamin C in rabbits. Bas J Vet Res
2016; 14:110-24.
192. Mudatsir M, Yufika A, Nainu F, Frediansyah A, Megawati D. Antiviral activity of ivermectin against SARS-
CoV-2: an old-fashioned dog with a new trick- Literature review. Sci Pharm 2020; 88:36.
193. Carvallo H, Hirsch R, Farinella ME. Safety and efficacy of the combined use of Ivermectin, dexamethasone,
enoxaparin and aspirin against COVID-19. medRxiv 2020.
194. Heaney RP, Armas LA, Shary JR, Bell NH, Binkley N, Hollis BW. 25-hydroxylation of vitamin D3: relation to
circulating vitamin D3 under various input conditions. Am J Clin Nutr 2008; 87:1738-42.
195. Murai IH, Fernandes AL, Sales LP, Pinto AJ, Goessler KF. Effect of vitamin D3 supplementaion vs placebo
on hospital length of stay in patients with severe COVID-19: A multicenter, double-blind, randomized
controlled trial. medRxiv 2020.
196. Menezes RR, Godin AM, Rodrigues FF, Coura GM, Melo IS, Brito AM. Thiamine and riboflavin inhibit
production of cytokines and increase the anti-inflammatory activity of a corticosteroid in a chronic model
of inflammation induced by complete Freund's adjuvant. Pharmacological Reports 2020; 69:1036-43.
197. Vatsalya V, Li F, Frimodig J, Gala KS, Srivastava S, Kong M. Therapeutic prospects for Th-17 cell immune
storm syndrome and neurological symptoms in COVID-19: Thiamine efficacy and safety, In-vitro evidence
and pharmacokinetic profile. medRxiv 2020.
198. Mallat J, Lemyze M, Thevenin D. Do not forget to give thiamine to your septic shock patient! J Thorac Dis
2016; 8:1062-6.
199. Moskowitz A, Donnino MW. Thiamine (vitamin B1) in septic shock: a targeted therapy. J Thorac Dis 2020;
12 (suppl 1):S78-S83.
200. Woolum JA, Abner EL, Kelly A, Thompson Bastin ML, Morris PE, Flannery AH. Effect of thiamine
administration on lactate clearance and mortality in patients with septic shock. Crit Care Med 2018;
46:1747-52.

Page 33 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


201. Marik PE. Thiamine: An essential component of the metabolic resuscitation protocol. Crit Care Med 2018;
46:1869-70.
202. Lee CY, Jan WC, Tsai PS, Huang CJ. Magnesium sulfate mitigates acute lung injury in endotoxemia rats. J
Trauma 2011; 70:1177-85.
203. Salem M, Kasinski N, Munoz R, Chernow B. Progressive magnesium deficiency inceases mortality from
endotoxin challenge:Protective effects of acute magnesium replacement therapy [abstract]. Crit Care Med
1995;A260.
204. Jiang P. Does hypomagnesemia impact on the outcome of patients admitted to the intensive care unit? A
systematic review and meta-analysis. Shock 2019; 47:288-95.
205. Spoorthi V, Sasank S. Utility of Ivermectin and Doxycycline combination for the treatment of SARS-CoV-2.
International Archives of Integrated Medicine 2020; 7.
206. Cloutier N, Allaeys I, Marcoux G, Machius KR, Mailhot B. Platelets release pathogenic serotonin and return
to circulation after immune complex-mediated sequestration. PNAS 2018;E1550-E1559.
207. Calfee CS, Delucchi KL, Sinha P, Matthay MA, Hackett J, Shankar-Hari M. Acute respiratory distress
syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised
controlled trial. Lancet Resp Med 2018; 6:691-8.
208. Zhang XJ, Qin JJ, Cheng X et al. In-hospital use of statins is associated with a reduced risk of mortality
among individuals with COVID-19. Cell Metabolism 2020.
209. Rodriguez-Nava G, Trelles-Garcia DP, Yanez-Bello MA, Chung CW. Atorvastatin associated with decreased
hazard for death in COVID-19 patients admitted to an ICU: a retrospective cohort study. Crit Care 2020;
24:429.
210. Gupta A, Madhavan MV, Poterucha TJ, DeFilippis EM, Hennessey JA. Association between antecedent
statin use and decreased mortality in hospitalized patients with COVID-19. Research Square 2020.
211. Kow CS, Hasan SS. Meta-analysis of effectiveness of statins in patients with severe COVID-19. Am J Cardiol
2020.
212. Tan WY, Young BE, Lye DC, Chew DE, Dalan r. Statin use is assocaited with lower disease severity in
COVID-19 infection. Nature Research 2020.
213. Oldenburg CE, Doan T. Azithromycin for severe COVID-19. Lancet 2020.
214. Futado RH, Berwanger O, Fonseca HA, Correa TD, Ferraz LR, Lapa MG. Azithromycin in addition to
standard of care versus standard of care alone in the treatment of patients admitted to the hospital with
severe COVID-19 in Brazil (COALITION II): a randomised trial. Lancet 2020.
215. Agarwal A, Mukherjee A, Kumar G, Chatterjee P, Bhatnager T. Convalescent plasma in the management of
moderate covid-19 in adults in India: open label phase II multicentre randomised controlled trial (PLACID
Trial). BMJ 2020; 371:m3939.
216. Simonovich VA, Pratx LD, Scibona P, Beruto MV, Vallone MG. A randomized trial of convalescent plasma in
COVID-19 severe pneumonia. N Engl J Med 2020.
217. Rosas IO, Brau N, Waters M et al. Tocilizumab in hospitalized patients with COVID-19 pneumonia. medRxiv
2020.
218. Hermine O, Mariette X, Tharaux PL, Resche-Rignon M, Porcher R. Effect of tocilizumab vs usual care in
adults hospitalized with COVID-19 and moderate or severe pneumonia.A randomized Clinical Trial. JAMA
Intern Med 2020.
219. Stone JH, Frigault MJ, Sterling-Boyd NJ, Fernandes AD, Harvey FL. Efficacy of tocilizumab in patients
hospitalized with Covid-19. N Engl J Med 2020.
220. Salvarani C, Dolci G, Massari M, Merlo DF, Cavuto S. Effect of tocilizumab vs standard care on clinical
worsening in patients hospitalized with COVID-19 pneumonia. A randomized clinical trial. JAMA Intern
Med 2020.
221. Bassetti M, Kollef MH, Timsit JF. Bacterial and fungal superinfections in critically ill patients with COVID-
19. Intensive Care Med 2020.
222. Xu Q, Wang T, Quin X, Zha L. Early awake prone position combined with high-flow nasal oxygen therapy in
severe COVID-19; a case series. Crit Care 2020; 24:250.
223. Elharrar X, Trigui Y, Dois AM, Touchon F. Use of prone positioning in nonintubated patients with COVID-19
and hypoxemic acute respiratory failure. JAMA 2020.
224. Keith P, Day M, Perkins L, Moyer L, Hewitt K, Wells A. A novel treatment approach to the novel
coronavirus: an argument for the use of therapeutic plasma exchange for fulminant COVID-19. Crit Care
2020.

Page 34 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


225. Keith P, Wells AH, Hodges J, Fast SH. The therapeutic efficacy of adjunct therapeutic plasma exchange for
septic shock with multiple organ failure: A single center experience. Crit Care 2020; 24:518.
226. Busund R, Koukline V, Utrobin U, Nedashkovsky E. Plasmapheresis in severe sepsis and septic shock: a
prospective, randomised, controlled trial. Intensive Care Med 2002; 28:1434-9.
227. Morath C, Weigand MA, Zeier M, Speer C, Tiwari-Heckler S. Plasma exchange in critically ill COVID-19
patients. Crit Care 2020; 24:481.
228. Khamis F, Al-Zakwani I, Al Hashmi S, Al Dowaiki S, Al Bahrani M. Therapeutic plasma exchange in adults
with severe COVID-19 infection. Int J Infect DIs 2020.
229. Fernandez J, Gratacos-Gines J, Olivas P, Costa M, Nieto S, Mateo D. Plasma exchange: An effective rescue
therapy in critically ill patients with Coronavirus Disease 2019 infection. Crit Care Med 2020.
230. Gucyetmez B, Atalan HK, Sertdemir I, Cakir U, Telci L. Therapeutic plasma exchange in patients with
COVID-19 pneumonia in intensive care unit: a retrospective study. Crit Care 2020; 24:492.
231. Poor HD, Ventetuolo CE, Tolbert T, Chun G, Serrao G. COVID-19 critical illness pathophysiology driven by
diffuse pulmonary thrombi and pulmonary endothelial dysfuncion responsive to thrombolysis. medRxiv
2020.
232. Wang J, Najizadeh N, Moore EE, McIntyre RC, Moore PK, Veress LA. Tissue plasminogen activator (tPA)
treatment for COVID-19 associated respiratory distress syndrome (ARDS): A case series. J Thromb
Haemost 2020.
233. Henry MB, Lippi G. Poor survival with extracorporeal membrane oxygenation in acute respiratory distress
syndrome (ARDS) due to coronavirus disease 2019 (COVID-19): Pooled analysis of early reports. J Crit Care
2020; 58:27-8.
234. Abrams D, Lorusso R, Vincent JL, Brodie D. ECMO during the COVID-19 pandemic: when is it unjustified.
Crit Care 2020; 24:507.
235. Barbaro RP, MacLaren G, Boonstra PS, Iwashyna TJ, Slutsky AS. Extracorporeal membrane oxygenation
support in COVID-19: an international cohort study of the Extracorporeal Life Support Organization
registry. Lancet 2020.
236. Abou-Arab O, Huette P, Debouvries F, Dupont H, Jounieaux V. Inhaled nitric oxide for critically ill Covid-19
patients: a prospective study. Crit Care 2020; 24:645.
237. Bagate F, Tuffet S, Masi P, Perier F, Razazi K. Rescue thearpy with inhaled nitric oxide and almitrine in
COVID-19 patients with severe acute respiratory distress syndrome. Ann Intensive Care 2020.
238. Caplan M, Goutay J, Bignon A, Jaillette E, Favory R. Almitrine infusion in severe acute respiratory
syndrome coronavirus-2 indued acute respiratory distress syndrome: A single-center observational study.
Crit Care Med 2020.
239. Payen D. Coronavirus disease 2019 acute respiratory failure: Almitrine drug resuscitaion or resuscitating
patients by almitrine? Crit Care Med 2020.
240. Zeng QL, Yu ZJ, Gou JJ, Li GM. Effect of convalescent plasma therapy on viral shedding and survival in
COVID-19 patients. Clin Infect Dis 2020.
241. Li L, Zhang W, Hu Y, Tong X, Zeng S, Yang J. Effect of convalescent plasma therapy on time to clinical
improvement in patients with severe and life-threatening COVID-19. A randomized clinical trial. JAMA
2020; 324:460-70.
242. Fleming AB, Raabe V. Current studies of convalescent plasma therapy for COVID-19 may underestimate
risk of antibody-dependent enhancement [letter]. J Clin Virol 2020; 127:104388.
243. Duan K, Liu B, Li C, Zhang H. Effectiveness of convalescent plasma therapy in severe COVID-10 patients.
PNAS 2020.
244. Cerutti A, Chen K, Chorny A. Immunoglobulin responses at the mucosal interface. Annu Rev Immunol
2011; 29:273-93.
245. Jacobs JJ. Neutralizing antibodies mediate virus-immue pathology of COVID-19. Med Hypotheses 2020;
143:109884.
246. Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor
Febratinib. J Microbiol Immunol Infect 2020.
247. Favalli EG, Biggioggero M, Maioli G, Caporali R. Baricitinib for COVID-19: a suitable treatment? Lancet
Infect Dis 2020.
248. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm
syndromes and immunosuppression. Lancet 2020; 395:1033-4.

Page 35 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


249. Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR. Baricitinib plus remdesivir for hospitalized
adults with COVID-19. N Engl J Med 2020.
250. Seifirad S. Pirfenidone: A novel hypothetical treatment for COVID-19. Medical Hypotheses 2020;
144:11005.
251. Saba A, Vaidya PJ, Chavhan VB, Achlerkar A, Leuppi J. Combined pirfenidone, azithromycin and
prednisolone in post-H1N1 ARDS pulmonary firbosis. Sarcoidosis Vasc Diffuse Lung Dis 2018; 35:85-90.
252. Spagnolo P, Balestro E, Aliberti S, Cocconcelli E, Biondini D, Casa GD. Pulmonary fibrosis secondary to
COVID-19: a call to arms? Lancet Resp Med 2020; 8:750-2.
253. George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and COVID-19: the potential role for antibibrotic
therapy. Lancet Resp Med 2020; 8:807-15.
254. Brouwer WP, Duran S, Kuijper M, Inc C. Hemoadsorption with CytoSorb shows a decreased observed
versus expected 28-day all-cause mortality in ICU patients with septic shock: a propensity-score-weighted
retrospective study. Crit Care 2019; 23:317.
255. Villa G, Romagnoli S, De Rosa S, Greco M, Resta M. Blood purification therapy with a hemodiafilter
featuring enhanced adsorptive properties for cytokine removal in patients presenting COVID-19: a pilot
study. Crit Care 2020; 24:605.
256. Bryce C, Grimes Z, Pujadas E et al. Pathopysiology of SARS-CoV-2: targeting of endothelial cells renders a
complex disease with thrombotic microangiopathy and aberrant immune response. The Mount Sinai
COVID-19 autopsy experience. medRxiv 2020.
257. Slaats J, ten Oever J, van de Veerdonk FL, Netea MG. Il-1B/Il-6/CRP and IL-18/ferritin: Distinct
inflammatory programs in infections. PLos Pathog 2016; 12:e1005973.
258. Colafrancesco S, Alessandri C, Conti F, Priori R. COVID-19 gone bad: A new character in the spectrum of
the hyperferritinemic syndrome? Autoimmunity Reviews 2020; 19:102573.
259. Giamarellos-Bouboulis EJ, Netea MG, Rovina N et al. Complex immune dysregulation in COVID-19 patients
with severe respiratory failure. Cell Host & Microbe 2020.
260. McGonagle D, Sharif K. The role of cytokines including interleukin-6 in COVID-19 induces pneumonia and
macrophage activation syndrome-like disease. Autoimmunity Reviews 2020.
261. Kyriazopoulou E, Leventogiannis K, Norrby-Teglund A, Dimopoulos G. Macrophage activation-like
syndrome: an immunological entity associated with rapid progression to death in sepsis. BMC Medicine
2017; 15:172.
262. Tan C, Huang Y, Shi F, Tan K, Ma Q, Chen Y. C-reactive protein correlates with computed tomographic
findings and predicts severe COVID-19 early. J Med Virol 2020; 92:856-62.
263. Howell AP, Parrett JL, Malcom DR. Impact of high-dose intravenous vitamin C for treatment of sepsis on
point-of-care blood glucose readings. J Diabetes Sci Technol 2019.
264. Stephenson E, Hooper MH, Marik PE. Vitamin C and Point of Care glucose measurements: A retrospective,
Observational study [Abstract]. Chest 2018; 154 (suppl.):255a.
265. Brosnahan SB, Bhatt A, Berger JS, Yuriditsky E, Iturrate E. COVID-19 pneumonia hospitalizations followed
by re-presentation for presumed thrombotic event. Chest 2020.
266. Spyropoulos AC, Lipardi C, Xu J, Peluso C, Spiro TE. Modified IMPROVE VTE Risk Score and elevated D-
Dimer identify a high venous thromboembolism risk in acutely ill medical population for extended
thromboprophylaxis. TH Open 2020; 4:e59-e65.
267. Kunutsor SK, Seidu S, Blom AW, Khunti K. Serum C-reactive protein increases the risk of venous
thromboembolism: a prospective study and meta-analysis of published prospective evidence. Eur J
Epidemiol 2017; 32:657-67.
268. Carfi A, Bernabei R, Landi F. Persistent symptoms in patients after acute COVID-19. JAMA 2020.
269. Prescott HC, Girard TD. Recovery from Severe COVID-19. Leveraging the lessons of survival from sepsis.
JAMA 2020.
270. Greenhalgh T, Knight M, A'Court C, Buxton M, Husain L. Management of post-acute Covid-19 in primary
care. BMJ 2020.
271. Chopra V, Flanders SA, O'Malley M. Sixty-day outcomes among patients hospitalized with COVID-19. Ann
Intern Med 2020.
272. Mandal S, Barnett J, Brill SE, Brown JS, Hare SS. 'Long-COVID': a cross-sectional study of persisting
symptoms, biomarker and imaging abnormalities following hospitalization for COVID-19. Thorax 2020.
273. Michelen M, Manoharan L, Elkheir N, Cheng V, Dagens D, Hastie C. Characterising long-term covid-19: a
rapid living systematic review. medRxiv 2020.

Page 36 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


274. Lu Y, Li X, Geng D, Mei N, Wu PY, Huang CC. Cerebral micro-structutal changes in COVID-19 patients - An
MRI-based 3-month follow-up study. EClinicalMedicine 2020.
275. Riche F. Protracted immune disorders at one year after ICU discharge in patients with septic shock. Crit
Care 2018; 22:42.
276. Andreakos E, Papadaki M, Serhan CN. Dexamethasone, pro-resolving lipid mediators and resolution of
inflammation in COVID-19. Allergy 2020.
277. Dalli J, Chiang N, Serhan CN. Elucidation of novel 13-series resolvins that increase with atorvastatin and
clear infections. Nat Med 2015; 21:1071-5.
278. Aguirre-Chang G, Castillo Saavedra E, Yui Cerna M, Trugjillo Figuerdo A. Post-acute or prolonged COVID-
19: Treatment with ivermectin for patients with persistent, or post-acute symptoms. Research Square
2020.
279. Gao J, Zheng P, Jia Y, Chen H. Mental health problems and social media exposure during COVID-19
outbreak. PloS ONE 2020; 15:e0231924.
280. Pennycook G, McPhetres J, Zhang Y, Lu JG, Rand DG. Fighting COVID-19 misinformation on Social Media:
Experimental Evidence fo a Scalable Accuracy-Nudge Intervention. Psychological Science 2020; 31:770-80.
281. Kurcicka L, Lauer SA, Laeyendecker O, Boon D, Lessler J. Variation in false-negative rate of reverse
transcriptase polmerase chain reacion-based SARS-CoV-2 tests by time since exposure. Ann Intern Med
2020; 173:262-7.
282. Cheng HY, Jian SW, Liu DP, Huang WT, Lin HH. Contact tracing assessment of COVI-19 transmission
dynamics in Taiwan and risk at different exposure periods before and after symptom onset. JAMA Intern
Med 2020; 180:1156-63.
283. Zhao J, Yang Y, Huang H, Li D, Gu D. Relationship between ABO blood group and the COVID-19
susceptibility. medRxiv 2020.
284. Banerjee A, Pasea L, Harris S, Gonzalez-Izquierdo A. Estimating excess 1-year mortality associated with the
COVID-19 pandemic according to underlying conditions and age: a population-based cohort study. Lancet
2020; 395:1715-25.
285. Goren A, Vamo-Galvan S, Wambier CG, McCoy J. A preliminary observation: Male pattern hair loss among
hospitalized COVID-19 patients in Spain- A potential clue to the role of androgens in COVID-19 severity. J
Cosmetic Dermatol 2020.
286. Huang C, Wang Y, Li X et al. Clinical features of patients infected with 2019 novel coronavirus in
Wuhan,China. Lancet 2020; 395:497-506.
287. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J. Clinical characteristics of Coronavirus disease 2019 in China. N
Engl J Med 2020.
288. von der Thusen J, van der Eerden M. Histopathology and genetic susceptibility in COVID-19 pneumonia.
Eur J Clin Invest 2020.
289. Sweeney TE, Liesenfeld O, Wacker J, He YD, rawling D, Remmel M. Validation of inflammopathic, adaptive,
and coagulopathic sepsis endotypes in Coronavirus disease 2019. Crit Care Med 2020.
290. Tartof SY, Qian L, Hong V, Wei R, Nadjafi RF, Fischer H. Obesity and mortality among patients diagnosed
with COVID-19: Results from an integrated health care organization. Ann Intern Med 2020.
291. Pujadas E, Chaudhry F, McBride R, Richter F, Zhao S. SARS-CoV-2 viral load predictes COVID-19 mortality.
Lancet Resp Med 2020.
292. Akbar AN, Gilroy DW. Aging immunity may exacerbate COVID-19. Science 2020; 369.
293. Zhang Q, Bastard P, Liu Z, Le Pen J, Chen J, Korol C. Inborn errors of type I IFN immunity in patients with
life-threatening COVID-19. Science 2020.
294. Zhou Y, Fu B, Zheng X, Wang D, Zhao C. Pathogenic T cellls and inflammatory monocytes incite
inflammatory storm in severe COVID-19 patients. Natl Sci Rev 2020; 7:998-1002.
295. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S. Imbalanced host response to SARS-CoV-2 drives
development of COVID-19. Cell 2020.
296. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z. Clinical course and risk factor for mortality of adult inpatients with
COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020.
297. Giamarellos-Bourboulis EJ, Netea MG, Rovina N, Akinosoglou K. Complex immune dysregulation in COVID-
19 patients with severe respiratory failure. medRxiv 2020.
298. Qin C, Zhou L, Hu Z, Zhang S. Dysregulation of the immune response in patiens with COID-19 in Wuhan,
China. Lancet Infect Dis 2020.

Page 37 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


299. Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. The cytokine release syndrome (CRS) of severe COVID-19 and
interleukin-6 receptor (IL-6R) antagonsit Tocilizumab may be the key to reduce the mortality. Int J
Antimicrob Agents 2020.
300. Ye Q, Wang B, Mao J. The pathogenesis and treatment of the "Cytokine Storm" in COVID-19. J Infection
2020.
301. Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science 2020.
302. Tay MZ, Poh CM, Renia L, MacAry PA. The trinity of COVID-19: immunity, inflammation and intervention.
Nature Reviews 2020; 20:363-74.
303. Leisman DE, Deutschman CS, Legrand M. Facing COVID-19 in the ICU: vascular dysfunction, thrombosis,
and dysregulated inflammation. Intensive Care Med 2020; 46:1105-8.
304. Teuwen LA, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nature Reviews 2020.
305. Varga Z, Flammer AJ, Steiger P, Habrecker M, Andermatt R, Zinkernagel AS. Endothelial cell infection and
endotheliitis in COVID-19. Lancet 2020.
306. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T. Pulmonary vascular endothelialitis,
Thrombosis, and Angiogenesis in COVID-19. N Engl J Med 2020; 383:120-8.
307. Kory P, Kanne JP. SARS-CoV-2 organizing pneumonia:"Has there been a widespread failure to identify and
treat this prevalent condition in COVID-19?'. BMJ Open Resp Res 2020; 7:e000724.
308. Torrealba JR, Fisher S, Kanne JP, Butt YM, Glazer C, Kershaw C. Pathology-radiology correlation of
common and uncommon computed tomographic patterns of organizing pneumonia. Human Pathology
2018; 71:30-40.
309. Kanne JP, Little BP, Chung JH, Elicker BM. Essentials for radiologists on COVID-19: an Update-Radiology
Scientific Expert Panel. Radiology 2020.
310. Copin MC, Parmentier E, Duburcq T, Poissy J, Mathieu D. Time to consider histologic pattern of lung injury
to treat critically ill patietns with COVID-19 infection [letter]. Intensive Care Med 2020.
311. Gattinoni L, Chiumello D, Caironi P, Busana M, Romitti F, Brazzi L. COVID-19 pneumonia: different
respiratory treatment for different phenotypes? Intensive Care Med 2020; 46:1099-102.
312. Chiumello D, Cressoni M, Gattinoni L. Covid-19 does not lead to a "typical" Acute Respiratory Distress
syndrome. Lancet 2020.
313. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia: ARDS or not? Crit Care 2020; 24:154.
314. Gattinoni L, Pesenti A. The concept of "baby lung". Intensive Care Med 2005; 31:776-84.
315. Patel AN, Desai SS, Grainger DW, Mehra MR. Usefulness of ivermectin in COVID-19 illness. medRxiv 2020.
316. Jeronimo CM, Farias ME, Almeida FF, Sampaio VS, Alexandre MA, Melo GC. Methylprednisolone as
adjunctive therapy for patients hospitalized with COVID-19 (Metcovid): A ramdomised, double-blind,
phase IIb, placebo-controlled trial. Clin Infect Dis 2020.
317. Carsana L, Sonzogni A, Nasr A, Rossi RS, Pellegrinelli A, Zerbi P. Pulmonary post-mortem findings in a large
series of COVID-19 cases from Northern Italy. medRxiv 2020.
318. Menter T, Haslbauer JD, Nienhold R, Savic S, Hopfer H. Post-mortem examination of COVID19 patients
reveals diffuse alveolar damage with severe capillary congestion and variegated findings of lungs and
other organs suggesting vascular dysfunction. medRxiv 2020.
319. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C. Pathological findings of COVID-19 associated with acute
respiratory distress syndrome. Lancet Resp Med 2020.
320. Tobin MJ, Laghi F, Jubran A. Why COVID-19 silent hypoxemia is baffling to physicians. Am J Respir Crit Care
Med 2020.
321. Schurink B, Roos E, Radonic T, Barbe E, Bouman CS. Viral presence and immunopathology in patients with
lethal COVID-19: a prospective autopsy cohort study. Lancet Microbe 2020.
322. Buijsers B, Yanginlar C, Maciej-Hulme ML, de Mast Q. Beneficial non-anticoagulant mechanisms
underlying heparin treatment of COVID-19 patients. EBioMedicine 2020.
323. Kim SY, Jin W, Sood A, Montgomery DW, Grant OC, Fuster MM. Characterization of heparin and severe
acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) spike glycoprotein binding interactions.
Antiviral Res 2020; 181:104873.
324. Clausen TM, Sandoval DR, Spliid CB et al. SARS-CoV-2 infection depends on cellular heparan sulphate and
ACE2. bioRxiv 2020.
325. Huang X, Han S, Liu x, Wang T, Xu H. Both UFH and NAH alleviate shedding of endothelial glycocalyx and
coagulopathy in LPS-induced sepsis. Exp Thera Med 2020; 19:913-22.

Page 38 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


326. Buijsers B, Yanginlar C, de Nooijer A, Grondman I, Jonkman I, Rother N. Increased plasma heparanase
activity in COVID-19 patients. medRxiv 2020.
327. May JM, Qu ZC. Ascorbic acid prevents oxidant-induced increases in endothelial permeability. Biofactors
2011; 37:46-50.
328. Utoguchi N, Ikeda K, Saeki K et al. Ascorbic acid stimulates barrier function of cultured endothelial cell
monolayer. Journal of Cellular Physiology 1995; 163:393-9.
329. Han M, Pendem S, Teh SL, Sukumaran DK, Wu F, Wilson JX. Ascorbate protects endothelial barrier
function during septic insult: Role of protein phosphatase type 2A. Free Radic Biol Med 2010; 48:128-35.
330. Elenkov IJ. Glucocorticoids and the Th1/Th2 balance. Ann N Y Acad Sci 2004; 1024:138-46.
331. Shodell M, Siegal FP. Corticosteroids depress INF-alpha-producing plasmacytoid dentritic cells in human
blood. J Allergy Clin Immunol 2001; 108:446-8.
332. Thomas BJ, Porritt RA, Hertzog PJ, Bardin PG. Glucocorticosteroids enhance replication of respiratory
viruses: effect of adjuvant interferon. Scientific Reports 2014; 4:7176.
333. Singanayagam A, Glanville N, Girkin JL, Ching YM, Marcellini A. Corticosteroid suppression of antiviral
immunity increases bacterial loads and mucus production in COPD exacerbations. Nature
Communications 2018; 9:2229.
334. Salton F, Confalonieri P, Santus P et al. Prolonged low-dose methylprednisolone in patients with severe
COVID-19 pneumonia. medRxiv 2020.
335. Braude AC, Rebuck AS. Prednisone and methylprednisolone disposition in the lung. Lancet 1983;995-7.
336. Draghici S, Nguyen TM, Sonna LA et al. COVID-19: disease pathways and gene expression chnages predict
methylprednisolone can improve outcome in severe cases. Nature Reviews 2020.
337. Chen L, Li J, Luo C, Liu H, Xu W, Chen G. Binding interaction of quercetin-3-B-galactoside and its synthetic
derivatives with SARS-CoV 3CLpro: Structure-activity relationship studies revela salient pharmacophore
features. Bioorganic & Medicinal Chemistry 2020; 14:8295-306.
338. Ono K, Nakane H. Mechanisms of inhibition of various cellular DNA and RNA polymerases by several
flavonoids. J Biochem 1990; 108:609-13.
339. Kaul TN, Middleton E, Pgra PL. Antiviral effects of flavonoids on human viruses. J Med Virol 1985; 15:71-9.
340. Shinozka K, Kikuchi Y, Nishino C, Mori A, Tawata S. Inhibitory effect of flavonoids on DNA-dependent DNA
and RNA polymerases. Experientia 1988; 44:882-5.
341. Martin JH, Crotty S, Warren P. Does an apple a day keep the doctor away because a phytoestrogen a day
keeps the virus at bay? A review of the anti-viral properties of phytoestrogens. Phytochemistry 2007;
68:266-74.
342. Smith M, Smith JC. Repurposing therapeutics for COVID-19: Supercomputer-based docking to the SARS-
CoV-2 viral spike protein and viral spike protein-human ACE2 interface. ChemRxiv 2020.
343. Leyva-Lopez N, Gutierrez-Grijalva EP, Ambriz-Perez D. Flavonoids as cytokine modulators: A possible
therapy for inflammation-related diseases. Int J Mol Sci 2016; 17:921.
344. Nair MP, Kandaswami C, Mahajan S, Chadha KC, Chawda R, Nair H. The flavonoid, quercetin, differentially
regulates Th-1 (INF) and Th-2 (IL4) cytokine gene expression by normal peripheral blood mononuclear
cells. Biochimica et Biophysica Acta 2020; 1593:29-36.
345. Dabbagh-Bazarbachi H, Clergeaud G, Quesada IM, Ortiz M, O'Sullivan CK. Zinc ionophore activity of
Quercetin and Epigallocatechin-gallate:From Hepa 1-6 cells to a liposome model. J Agric Food Chem 2014;
62:8085-93.
346. Rhodes J, Dunstan F, Laird E, Subramanian S, Kenny RA. COVID-19 mortality inceases with northerly
latitude after adjustment for age suggesting a link with ultraviolet and vitamin D. BMJ Nutrition,
Prevenion & Health 2020.
347. Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with decreased mortality
in severe coronavirus disease 2019 with coagulopathy. medRxiv 2020.
348. Sardu C, Gambardella J, Morelli MB, Wang X. Is COVID-19 an endothelial disease? Clinical and basic
evidence. medRxiv 2020.
349. World Health Organization: Coronavirus Disease 2019 (COVID-19): Situation Report -54 (14th March
2020). https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200314-sitrep-54-
covid-19.pdf . 2020. 7-9-2020.
350. Clinical management of COVID-19. Interim guidance. 27th May 2020.
https://www.who.int/publications/i/item/clinical-management-of-covid-19 WHO/2019-
nCoV/clinical/2020.5 . 2020. World Health Organization. 7-10-2020.

Page 39 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17


351. Yam LY, Lau AC, Lai FY, Shung E, Chan J. Corticosteroid treatment of severe acute respiratory syndrome in
Hong Kong. J Infection 2007; 54:28-39.
352. Siemieniuk RA, Bortoszko JJ, Ge L, Zeraatkar D, Izcovich A. Drug treatments for Covid-19: living systematic
review and network meta-analysis. BMJ 2020.
353. Saune PM, Bryce-Alberti M, Portmann-Baracco AS, Accinelli RA. Methylprednisolone pulse therapy: An
alternative management of severe COVID-19. Respiratory Medicine Case Reports 2020; 31:101221.
354. Fernandez-Cruz A, Ruiz-Antoran B, Gomez AM et al. Impact of glucocorticocoid treatment in SARS-CoV-2
infection mortality: A retrospective controlled cohort study. medRxiv 2020.
355. Corral-Gudino L, Bahamonde A, Arnaiz-Revillas F et al. GLUCOCOVID: A controlled trial of
methylprednisolone in adults hospitalized with COVID-19 pneumonia. medRxiv 2020.
356. Stauffer WM, Alpern JD, Walker PF. COVID-19 and dexamethasone. A potential strategy to avoid steroid-
related Strongyloides hyperinfection. JAMA 2020; 324:623-4.

Page 40 of 40 | EVMS – COVID-19 Management Protocol 2020-12-17

You might also like